Language selection

Search

Patent 2087008 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent Application: (11) CA 2087008
(54) English Title: NOVEL RECEPTOR-TYPE PHOSPHOTYROSINE PHOSPHATASE
(54) French Title: PHOSPHOTYROSINE-PHOSPHATASE DE RECEPTEUR
Status: Dead
Bibliographic Data
(51) International Patent Classification (IPC):
  • C12N 15/55 (2006.01)
  • C07K 14/705 (2006.01)
  • C12N 1/19 (2006.01)
  • C12N 1/21 (2006.01)
  • C12N 5/10 (2006.01)
  • C12N 9/16 (2006.01)
  • C12P 21/08 (2006.01)
  • C12Q 1/44 (2006.01)
  • G01N 33/566 (2006.01)
  • G01N 33/573 (2006.01)
  • G01N 33/577 (2006.01)
  • A61K 38/00 (2006.01)
  • C12Q 1/68 (2006.01)
(72) Inventors :
  • SCHLESSINGER, JOSEPH (Israel)
(73) Owners :
  • NEW YORK UNIVERSITY (United States of America)
(71) Applicants :
(74) Agent: OSLER, HOSKIN & HARCOURT LLP
(74) Associate agent:
(45) Issued:
(86) PCT Filing Date: 1991-07-11
(87) Open to Public Inspection: 1992-01-23
Examination requested: 1995-08-28
Availability of licence: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/US1991/004892
(87) International Publication Number: WO1992/001050
(85) National Entry: 1993-01-08

(30) Application Priority Data:
Application No. Country/Territory Date
551,270 United States of America 1990-07-11
654,188 United States of America 1991-02-26

Abstracts

English Abstract

2087008 9201050 PCTABS00010
A novel receptor-type protein tyrosine phosphatase protein or
glycoprotein and the DNA coding therefor is expressed in a wide
variety of mammalian tissues. Included in this family of proteins are
human R-PTPase-.alpha., human R-PTPase-.beta. and human
R-PTPase-gamma. The R-PTPase protein or glycoproteins may be produced by
recombinant means. Antibodies to the proteins, methods for measuring
the quantity of the proteins, methods for screening compounds,
such as drugs, which can bind to the proteins and inhibit or
stimulate their activity, are provided.


Claims

Note: Claims are shown in the official language in which they were submitted.


WO 92/01050 PCT/US91/04892
- 66 -

WHAT IS CLAIMED IS:

1. A human receptor type protein tyrosine
phosphatase (R-PTPase) protein or glycoprotein molecule
other than leucocyte common antigen (CD45) and leucocyte
common antigen-related protein (LAR), a functional
derivative thereof, or a homolog thereof in other mammals,
wherein, when said molecule is one which naturally occurs,
said molecule is substantially free of other proteins or
glycoproteins with which it is natively associated, said
naturally-occurring molecule being normally present in
mammalian liver, kidney and brain.

2. A molecule according to claim 1, which does
not occur in nature.

3. A molecule according to claim 1, which occurs
in nature and is substantially free of other proteins or
glycoproteins with which it is associated in natively
associated.

4. A molecule according to claim 1 designated
R-PTPase-.alpha. having the amino acid sequence shown in Figure 4,
or a functional derivative thereof.

5. A molecule according to claim 1 designated
R-PTPase-.beta. having the amino acid sequence shown in Figure 5,
or a functional derivative thereof.

6. A molecule according to claim 1 designated
R-PTPase-gamma having the amino acid sequence SEQ ID:NO. 3,
or a functional derivative thereof.

WO 92/01050 PCT/US91/04892
- 67 -

7. A DNA molecule encoding an R-PTPase protein
according to claim 1, or encoding a functional derivative
thereof, wherein, when said protein or said functional
derivative is one which naturally occurs, said DNA molecule
is substantially free of nucleotide sequences encoding
proteins with which said protein or said functional
derivative are natively associated.

8. A DNA molecule according to claim 7 which is
a cDNA sequence.

9. A DNA molecule according to claim 7 which is
a genomic DNA sequence.

10. A DNA molecule according to claim 7 having
the nucleotide sequence selected from the group consisting
of SEQ ID:NO. 4, SEQ ID:NO. 5, SEQ ID:NO. 6, or a functional
derivative of said sequences.

11. A DNA molecule according to claim 7 which is
an expression vehicle.

12. The DNA molecule of claim 11 wherein said
expression vehicle is a plasmid.

13. A prokaryotic host transformed with the DNA
molecule of claim 12.

14. A host according to claim 13 which is a
bacterium.

15. A eukaryotic host transfected with a DNA
molecule according to claim 11.

WO 92/01050 PCT/US91/04892
- 68 -

16. A host according to claim 15 which is a yeast
cell or a mammalian cell.

17. A process for preparing an R-PTPase protein
or glycoprotein according to claim 1, or a functional
derivative thereof, said process comprising:
(a) culturing a host capable of expressing said
protein under culturing conditions,
(b) expressing said protein; and
(c) recovering said protein from said culture.

18. A process according to claim 17 wherein said
host is a prokaryote.

19. A process according to claim 18 wherein said
host is a eukaryote.

20. An antibody specific for the protein or
glycoprotein of claim 1.

21. An antibody according to claim 20 which is
monoclonal.

22. A method for detecting the presence of a
nucleic acid sequence according to claim 7, or a nucleic
acid sequence encoding a mutant R-PTPase, in a subject
comprising:
(a) contacting a cell or an extract thereof from
said subject with an oligonucleotide probe
encoding at least a portion of said normal or
mutant R-PTPase under hybridizing conditions;
and
(b) measuring the hybridization of said probe to
the nucleic acid of said cell,

-69-

thereby detecting the presence of said nucleic acid
sequence.

23. The method of claim 22, additionally
comprising before step (a):
(c) selectively amplifying the amount of DNA of
said cell encoding said R-PTPase.

24. A method for detecting in a cell the presence
or measuring the quantity of an R-PTPase, comprising:
(a) contacting said cell or an extract thereof
with an antibody according to claim 20; and
(b) detecting the binding of said antibody to
said cell or extract thereof, or measuring
the quantity of antibody bound,
thereby determining the presence or measuring the quantity
of said R-PTPase protein or glycoprotein.

25. A method for identifying in a chemical or
biological preparation a compound capable of binding to an
R-PTPase protein, glycoprotein or derivative according to
claim 1, said method comprising:
(a) attaching said R-PTPase protein, glycoprotein
or derivative, or the ligand-binding portion
thereof to a solid phase matrix;
(b) contacting said chemical or biological
preparation with said solid phase matrix
allowing said compound to bind, and washing
away any unbound material; and
(c) detecting the presence of said compound bound
to said solid phase.

WO 92/01050 PCT/US91/04892
- 70 -

26. A method for isolating from a complex mixture
a compound capable of binding to an R-PTPase protein,
glycoprotein or functional derivative according to claim 1,
comprising:
(a) attaching said R-PTPase or functional
derivative, or the ligand-binding portion
thereof, to a solid phase matrix;
(b) contacting said complex mixture with said
solid phase matrix allowing said compound to
bind, and washing away any unbound material;
and
(c) eluting said bound compound,
thereby isolating said compound.

27. A method for identifying a compound capable
of stimulating or inhibiting the enzymatic activity of an R-
PTPase comprising:
(a) contacting said compound with said R-PTPase
in pure form, in a membrane preparation, or
in a whole cell;
(b) incubating said mixture of step (a) for a
sufficient interval;
(c) measuring the enzymatic activity of said R-
PTPase;
(d) comparing said enzymatic activity to that of
said R-PTPase incubated without said
compound,
thereby determining whether said compound stimulates or
inhibits said activity.

Description

Note: Descriptions are shown in the official language in which they were submitted.


20~79~8
NOVEL Rl~CEPTOR--q~YPE P~OSPEIOTYROSINE PBOSPHATAS~
BACRGROI~ND OF T~I; INV~ITION
Field of the Invention
~ he invention in the field of biochemistry and
cell and molecular biology relates to novel receptor-type
protein tyrosine phosphatase proteins or glycoproteins,
termed R-PTPase-~, B and gamma, DNA coding therefor,
methods for production and identification of the proteins,
and methods for screening compounds capable of binding to
10 and inhibiting or stimulating PTPase enzymatic activity.

Description of the Backaround Art
The identification of several growth factor
receptors and retroviral oncogenes as tyrosine-specific
protein kinases indicated that protein phosphorylation on
15 tyrosine residues plays a key role in cellular growth
control. This notion has recently received support by the
observation that the level of tyrosine phosphorylation of
enzymes thought to play an important role in signal
transduction (such as phospholipase C) correlates with their
20 increased activity upon growth factor stimulation, thus ;
establishing a functional role for tyrosine phosphorylation `
~Ullrich, A., et al., Cell 61:203-212 (1990)).
The degree and pattern of phosphorylation of
tyrosine residues on cellular proteins are regulated by the
25 opposing activities of protein-tyrosine kinases (PTRases;
~TP:protein-tyrosine 0-phosphotransferase, EC 2.7.1.112) and
protein-tyrosine-phosphatases SPTPases; protein-tyrosine-
phosphate phosphohydrolase, EC 3.1.3.48). The structural
characteristics and evolution of PTKases as well as their
30 role in the regulation of cell growth have been reviewed
(Hunter, T., et al., Annu. Rev. Biochem. 54:897-930 (1985);
Ullrich, A., et al., sup~a).

.

WO92/01050 PCT/US91/~2
2~7~)0~


Tyrosine kinases comprise a discrete family of
enzymes having common ancestry with, but major differences
from, serine/threonine-specific protein kinases (Hanks, S.K.
et al., (lg88) Science 241, 42-52). The mechanisms leading
5 to changes in activity of tyrosine kinases are best
understood for receptor-type tyrosine kinases which have a
transmembrane topology (Ullrich, A. e~ al., supra). With
such kinases, the binding of specific ligands to the
extracellular domain of these enz~mes is thought to induce
10 their oligomerization leading to an increase in tyrosine
kinase activity and activation of the signal transduction
pathways (Ullrich, A. et al., supra). The importance of
this activity is supported by the knowledge that
dysregulation of kinase activity through mutation or over-
15 expression is a mechanism for oncogenic transformation(Hunter, T et al., supra; Ullrich, A. et al,, 1990, supra).
The protein phosphatases are composed of at least
two separate and distinct families (H~mter, T. Cell,
58:1013-1016 (1989)), the protein serine/threonine
20 phosphatases and the protein tyrosine phosphatases. This is
in contrast to protein kinases, which show clear sequence
similarity between serine/threonine-specific and tyrosine-
specific enzymes.
There appear to be two varieties of PTPase
25 molecules. The first group is comprised of small, soluble
enzymes that contain a single conser~ed phosphatase
catalytic domain, and include (1) placental PTPase lB
(Charbonneau, H. et al., Proc. Natl. Acad. Sci. 86:5252-5256
(1989); Chernoff, J. et al., Proc. Natl. Acad. Sci. USA
30 87:2735-2789 (1990)~, (2) T-cell PTPa,se (Cool, D.E. et al.,
~oc. Natl. Acad. Sci. USA 86:5257- 5261 (1989)), and (3)
rat brain PTPase (Guan, K., et al., P~oc. Natl. Acad. Sci.
USA, 87:1501- 1505 (1990).
The second group is made upl of the more complex,
35 receptor-linked PTPases, termed R PTPases, which are of high




:.: . . . . . ~ . ,
,

2V~ 7~8

molecular weight and contain two tandemly repeated conserved
domains separated by 56-57 amino acids. One example of R-
PTPases are t~e leukocyte common antigens (LCA) (Ralph,
S.J., EMBO J., 6:1251-1257 (1987); Charbonneau, H., et al.,
5 Proc. Natl. Acad. sci. US~, 85:7182~7186 (1988)). LCA, also
known as CD45, T200 and Ly-5 (reviewed in Thomas, N.L., Ann.
Rev. Immunol. 7:339-369 (19a9)) comprises a group of
membrane glycoproteins expressed exclusively in hemopoietic
(except late erythroid) cells, derived from a common gene by
10 alternative splicing events involving the amino terminus of
the proteins. Whereas the precise f~mction of CD45 is un-
known, many studies have implicated t:hese antigens in a
number of processes, including the ac:tivity of cytotoxic T
lymphocytes and natural killer cells, IL-2 receptor
15 expression, B-cell differentiation, and T lymphocyte
proliferation (Pingel, J.T. et a~., ~11 58:1055-1065
(1989)).
Other examples of R-PTPases are the LCA-related 1 -
protein, LAR (Streuli, M., et al., J. Exp. Med., 168:1523-
20 1530 (1988)), and the LAR-related Drosophila proteins DLAR
and DPTP (Streuli, M., et al~, Proc. Natl. Acad. ~ci. USA,
86:8698-8702 (1989)). Jirik et al. screened a cDNA library
derived from the human hepatoblastoma cell line, HepG2, with
a probe encoding the two PTPase doma:ins of LCA (FASEB J.
25 4:A2082 (1990), abstr. 2253) and discovered a cDNA clone
encoding a new R-PTPase, named He-PTP. The HePTP gene
appeared to be expressed in a variety of human and murine
cell lines and tissues. ;
While we are beginning to understand more about
30 the structure and diversity of the PTPases, much remains to
be learned about their cellular funct:ons. It has been
~uggested (Tonks, N.K., et al., Bioch~mistry, 27:8695-8701
(1988)) that the small, soluble PTPase enzymes may have a
"housekeeping" function. on the other hand, the R-PTPases
35 would be expected to be more restricted in their activities




'' , '',; ' ".' ' '' ''' " ' . ,','''' ~ ,'' '' ''," .' ' ' " '
': . ' .' - :, , . , , ' ',. ' .

~0 ~2/UIUSII PCr/USgl/04892
2~j~ J~

- 4 -
because of their location in the call membrane and their
potential regulation by extracellular ligands. Regarding
the role of LCA (cD4s) in T cells, it was found that T cell
clones deficient in the expression af LCA failed to
5 proliferate when stimulated by a spècific antigen or by
cross-linking of CD3 (Pingel, J.T., et al., supra). PTPase
cross-linking inhibits T cell receptor CD3-mediated
activation in human T cells (Kiener, P.A. et al., J.
Immunol. 14~:23-28 (1989)). The PTPase activity of LCA
10 plays a role in the activation of pp561ck, a lymphocyte-
specific PTKase (Mustelin, T., et al., Proc. Natl. Acad.
Sci. USA, 86:6302-6306 (1989); Ostergaard, H.L., et al.,
Proc. Natl. Acad. Sci. Us~, 86:8959-8~63 (1989)). These
authors hypothesized that the phosphatase activity of LCA
15 activates pp561ck by dephosphorylation of a C-terminal
tyrosine residue, which may, in turn, be related to T-cell
activation.
Using site-directed mutagenesis to determine
which of four conserved cysteines in ~CA (two per
20 phosphatase do~ain) was required for enzyme activity toward
artificial substrates, Streuli et al. (1989, sup~) found
that only one cysteine residue (residue 177 of LCA
phosphatase domain-l) of LCA was essential for activity,
indicating that, most likely, only the fir6t phosphatase
25 domain has enzymatic activity. However, the possibility
that the second domain can dephosphorylate a different
substrate was not excluded. ~ore recently, Streuli et. al.
(EMBo J2, 9:2399-2407 (~990)) determin~ed that the econd
conserved domain of LCA (and of LAR) lacked detectable
30 phosphatase activity but sequences within the domain could
influence substrate specificity.
In order to better understand and to be abl~ to
control phosphotyrosine metabolism, one must comprehend not
only the role of kinase activity, but also the action of
35 phosphatase enzymes as well. Elevation of cellular




;

'

20~7!)1~8


phosphotyrosine may occur through mec]lanisms not involving
the activation of a tyrosine kinase $tself. For instance,
expression of the v-crk oncogene, thol.lgh not a tyrosine
kinase itself, induces the phosphoryl~tion of tyrosine
5 residues through a poorly understood mechanism (Mayer, B.J.
et al. (1988) Nature 332, 272-275). ~otentially, such an
outcome could result from either mutation of the substrate
or through a general decrease in cel]ular phosphatase
activity, especially in view of the ns:~rmally high turnover
10 rate of cellular tyrosine-phosphate ~3efton, B.M. et al.
(1980) Cell 20, 807-816). The latter possibility is
suggested by the demonstration that t:yrosine phosphatase
inhibitors can "reversibly transform''' cells (Klarlund, J.K.
Cell 41: 707-717 (1985)). PTPases could therefore be viewed
15 as potential recessive oncogenes.
It is becoming clear that dlephosphorylation of
tyrosine can by itself function as an important regulatory
mechanism. Dephosphorylation of a C-1:er~inal tyrosine
residue stimulates tyrosine kinase ac1:ivity in the src-
20 family of tyrosine kinases (Hunter, T (1987) Cell 49, 1-4).
Tyrosine dephosphorylation has been suggested to be an
obligatory step in the mitotic activa1:ion of the MPF
(maturation promoting factor) kinase (Morla, A.O. et al.
(1989) Cell 58, 193-2~3). L~stly, mu1:ant analysis of
25 primitive eukaryotes has established c:rucial roles for -
serine phosphatase in cellular physiology (Cyert, M.S. et
al. (1989) Cell 57, 891-893). These observations point out
the need in the art for increasing our understanding of the
~echanisms that regulate tyrosine phosphatase activity.
It is clear in the art that further analysis of
structure-function relationships among these membrane
receptors are needed to gain important understanding of the
~echanisms of cell growth, differentiation, and oncogenesis.




: -., : - ,., , . .. ~,.. :. ,. ~ , . . . , -
.: -.. - .. . -. .. . . . . .
- :: ,: , . , . : - .
:- : , .. . ,., ~ : . : . .
, , . : - . :- - , . . . :,
: ' . ., : - '' : '
- : - - : . . ~ . :.
:: - ~ : . : , .
: : . . :: ' ,: . . '
: . . ,

L / ~ U ~ U~ I J I ~O~L
20~7~08


S~X~ARY 0~ THE INV~NTION
The inventors have conceiv~!d of a role for R-
PTPases in cellular c~ntrol mechanisms, both as potential
anti-oncogenes, and as effectors in ~ newly discovered
5 mechanism of transmembrane signalling. They therefore
undertook a search for an R-PTPase potentially involved in
such processes, and describe herein the identification of a
novel, widely expressed member of the R-PTPase family, which
has a transmembrane topology. Importantly, its
10 extracellular domain is unrelated to any other R-PTPase
heretofore described. The novel R-PTPase, in a m~nner
analogous to receptor tyrosine kinases, is subject to direct
regulation by a variety of different extracellular ligands.
The present invention thus provides a human
15 receptor-type protein tyrosine phosphatase (R-PTPase)
protein or glycoprotein molecule othe!r than leucocyte common
antigen (LCA or CD45) and leucocyte common antigen-related
protein (LAR), a functional derivative of the human ~-PTPase
or a homolog of the human R-PTPase in another mammalian
20 species. When the molecule is of natural origin, it is
substantially free o~ other proteins sr glycoproteins with
which it is natively associated. This naturally-occurring
molecule is normally present in mammalian liver, kidney and
brain. Alternatively, the R-PTPase molecule may not be of
25 natural origin, such as one prepared by chemical or
recombinant means.
The substantially pure R-FTPase protein or
glycoprotein of the invention may be produced by biochemical
purification of the glycoprotein of natural origin;
30 alternatively, the R-PTPase ~ay be produced by recombinant
means in prokaryotic or eukaryotic hosts.
In particular, the invention is directed to the
molecule R-PTPase-~ having the amino acid sequence of Figure
4, or a functional derivative thereof. In another
3~ embodiment, the invention is directed to human R-PTPase-~.




~: .


.., : . ' ' ~'
' ::

2~7~a8

-- 7 --
In yet another embodiment, the invention is directed to
human R-PTPase-gamma.
The inventio~ is further directed to a DNA
molecule consisting essentially of a nucleotide sequence
5 encoding ~-PTPase-~ of mouse or human origin, or R-PTPase-B
or R-PTPase-gamma, both of human origin, or a functional
derivative thereof, in the form of cDNA or genomic DNA. The
invention is further directed to the DNA sequence in the
form of an expression vehicle, as well as prokaryotic and
10 eukaryotic hosts transformed with the DNA.
Also included in the present invention is a
process for preparing an R-PTPase protein or glycoprotein of
this invention, or a functional derivative thereof,
comprising:
(a) culturing a host capable of ~xpressing the
protein under culturing conditions,
(b) expressing the protein; and
(c) recovering the protein from the culture.
The invention is directed to an antibody,
20 polyclonal, monoclonal, or chimeric, specific for the R-
PTPase-~ protein or glycoprotein.
The invention is also directed to a method for de-
tecting the presence of nucleic acid encoding a normal or
mutant R-PTPase in a subject comprising:
(a) contacting a cell or an extract thereof from
the subject with an oligonucleotide probe
encoding at least a portion of the normal or
mutant R-PTPase under hybridizing conditions;
and
(b) measuring the hybridization of the probe to
the nucleic acid of th~ cell, thereby
detecting the presence of the nucleic acid.
The DNA can be selectively amplified, using the polymerase
chain reaction, prior to a~say.




- . . . ~ . - . . . - .~
.:. . - ~- - . , : . :
.. .. ~ . .,. ~ .

. . : . . . . .. -

, ~ - , . ' : ': '

~ u l'~, l / Ui~Y l ~ Vq-SY ~
203'~0~8


The invention is further directed to a method for
detecting the presence, or measuring the quantity of an ~-
PTPase in cell or in a subject comprising:
(a) contacting said cell or an extract thereof
with an antibody specific for an epitope of
the R-PTPase; and
(b) detecting the binding of the antibody to the
cell or extract thereof, or measuring the
quantity of antibody bound,
10 thereby detecting the presence or measuring the guantity of
the R-PTPase.
The present invention is also directed t~ methods
for identifying and isolating a compound capable of binding
to an R-PTPase from a chemical or biological preparation
lS comprising:
(a) attaching the R-PTPase or the ligand-binding -~
portion thereof to a solid phase matrix;
(b) contacting the chemical or biological
preparation with the solid phase matrix
allowing the compound to bind, and washing
away any unbound material;
(c) detecting the presence of the compound bound
to the solid phase; and, for purposes of
isolation, : ~-
(d) elutiny the bound compound, thereby isolating
the compound.
Finally, the invention includes a method for
identifying a compound capable of stimulating or inhibiting
the enzymatic activity of a R-PTPase, comprising:
(a) contacting the compound with the R-PTPase in
pure form, in a ~embrane preparation, or in a
whole live or fixed cell; .
(b) incubating the mixture in step (a) for a
sufficient interval; :




: : ~ ~ ;, : , :

20~ 7~ 08


(c) measuring the enzymatic activity of the R-
PTPase;
(d) comparing the enzymatîc activity to that of
the R-PTPase incubated without the compound,
thereby determining whether the compound stimulates or
inhibits the activity.

BRIEF DESCRIPTION oP THE DRAWINGS

Figure 1 presents the predicted primary structure
of murine R-PTPase-~. Panel (a) shows the sequence of the
10 phage lambda-109 cDNA insert and predicted R-PTPase-
~protein sequence (the standard one letter amino acid code is
used). The initiation codon ATG is shown in italics, and
the stop codon is indicated by an asterisk. The putative
trans-membrane domain (amino acids 143 to 166) is u~derlined
15 as well as the potential N-linked glycosylation sites in the
extracellular domain. The borders of homology between the
tandemly repeated PTPase domains (I and II) are indicated by
square brackets. Cysteine residues conserved in the
catalytic domain of all known R-PTPases are also underlined.
20 Panel (b) shows a schematic structure of a lambda-109 cDNA
clone containing the R-PTPase-~ coding sequence. R-PTPase
domains I and II are indicated as black boxes, the
transmembrane domain is shaded. The start of the N-
terminally truncated PTP-DeltaC protein mentioned in Fiqure
2S 3, below is indicated by an arrow (at a~ino acid 214). The
positions of restriction sites used for generating nested
deletions for sequencing are indicated. Abbreviations: T~,
transmembrane domain; B, Ba~HI site; Bs, BstEII site; N,
NcoI site; Nd, NdeI site; P, PstI site; R, EcoRI site; S:
30 SacII site; St, StuI site.
Fiqure 2 is a diagram o~ a Northern blot showinq
expression of the ~urine R-PTPase-Q ~I~NA. 5 ~g of Poly A+
RNA from mouse tissues and cell lines was fractionated on

20~7~3~


-- 10 --
formaldehyde-c~ntaining agarose gels and subjected to
Northern analysis using as a probe the entire R-PTPase-
~cDNA. The positions of the 28S and 18S ribosomal RNA i5
indicated.
5 Lanes: 1, kidney; 2, lung; 3, heart; ~, stomach; 5, brain;
6, spleen; 7, liver; 8, NIH-3T3 fibroblast cell line
(Honegger, A.M. etTal. (1987) Cell 51, 199-209); 9, BAF
prepro-B lymphoid cell line (Palacios, R. et al. (1985) Cell
41, 727-734).
Figure 3 is a diagram showing results of PAGE of
immunoprecipitates of the murine R-P~'Pase-~ protein. Cos
cells were transiently transfected us;ing the DEAE-dextran
method with a negative control plasmîd (expression vector
pLSV without insert), with either pL5;V-PTP-~ (the same
15 expression vector containing the R-PTPase-~ cDNA), or with
the expression vector pLSVDeltaC, designed to express a
truncated R-PTPase-~ protein ~PTP-DeltaC, amino-acids 214-
794) from which the transmembrane andi extracellular domains
have been removed (an initiator methionine residue was
20 introduced at this position using site-directed
mutagenesis). After metabolic labelling with [35S]-
methionine, immunoprecipitation was performed using either
pre-immune serum (lanes 1 and 2) or with an antiserum (2A)
(lanes 3-8), raised against a synthetic peptide
25 corresponding to the C-terminus of the R-PTPase-~ protein in
the absence or presence of 100 ~g of the immunizing peptide.
Sizes of molecular weight markers are shown in kDa. The
arrow marks the position of the 130 kD R-PTPase-~ protein
(lane 5).
30 Lanes are: 1: pLSV, pre-immune serum; 2: pLSV-PTP-~, pre-
im~une serum; 3: pLSV, anti~erum 2A; 4: pLSV, antiserum 2A
in the presence of synthetic peptide; 5: pLSV-PTP-~,
antiserum 2A; 6: pLSV-PTP-~, antiseru~ 2A in the presence
of synthetic peptide; 7: pLSVDeltaC, ~ntiserum 2A; 8:




, ... - ~ . . ...

:- :

' :' '

w~ y~/u l u~u
2l)o 7~8


pLSVDeltaC, antiserum 2A in the presence of synthetic
peptide.
Figure 4 shows the structure of human R-PTPase-
~deduced from the sequence of cDNA clones.
(A) Composite restriction map [3615 base pairs (bp)] of
overlapping clones 31-4 and 27-1, which together contain the
entire coding region of human R-PTPase-~.
(B) Relative positions of clones 31-4 and 27-1. Both
strands of each clone were sequenced in their entirety by
10 using a series of oligonucleotide primers. The hatched
region in clone 31-4 corresponds to the fragment used as
probe for the Northern blot (Figure 6, below) as well as for
the chromosome assignment.
(C) Comparison of the amino acid sequences of human (line 1)
15 and mouse (line 2) R-PTPase-~. The single-letter amino acid
code is used. Only the differences are shown. The da~hed
line indicates a stretch of amino ac:ids not present in the
mouse sequence. The coding portion of human R-PTPase-~, and
its position relative to clones 31-4 and 27-1 (B), is shown
20 at the top. The following regions are designated: signal
peptide (I), extracellular domain with potential N-
glycosylation sites for the human protein underlined (II),
transmembrane ~III), juxtamembrane (IV), first phosphatase
domain (V), interdomain (VI), second phosphatase domain
25 (VII), C terminus (VIII).
Figure 5 shows a comparison of the amino acid
sequences of the first (A) and second (B) conserved
phosphatase of human R-PTPases LCA, a, B and gamma. CON is
the consensus sequence: a capital letter indicates complete
30 agreement, while a small letter indicates agreement amon~
two or three of the four sequences. A dash indicates lack
of oonsensus.
Figure 6 shows a gel pattern indicating relative
expres6ion of human R-PTPase-~ in various tissues and cell
35 lines, as determined by Northern blot hybridization with R-




- , .

~ I U~ '~ I / U~Y I ~ U4~
2~(~ 7~8


PTPase-~ probe (Upper) and B-actin probe (Lower). Total RNA
(five left lanes) or poly (A)+ RNA l~five right lanes)
samples from the indicated human ce:ll lines or tissues were
analyzed. A431 is a human epidermoid carcinoma cell line;
5 HEL is an erythroleukemia cell line î all other lanes
represent flash-frozen tissues samples (HUVEC - human
umbilical vein endothelial cells).
Figure 7 is a matrix diagr?lm which shows the
chromosomal localization of human R-~PTPase-~ based on
10 analysis of a panel of 17 rodent-human somatic cell hybrids.
A completely stippled box indicates that the hybrid
contained the chromosome indicated in the upper row; lower-
right stippling indicates presence oI` the long arm (or part
of the long arm, indicated by a smaller fraction of
15 stippling) of the chromosome; upper-left stippling indicates
presence of the short arm (or partial short arm) o~ the
chromosome; an open box indicates abs,ence of the chromosome.
The column for chromosome 20 is boldly outlined and stippled
to highlight correlation of presence of this chromosome (or
20 chromosome region) with the presence of the R-PTPase-~ gene.
The pattern of retention of the human R-PTPase-~ sequences
in the hybrids is shown at right (RPTP~): presence of the
gene is indicated by a "+" in a stip]pled box; absence of the
gene is indicated by a 11_~ in an open box.

2~ DESCRIPTION OP T~ PREF~RE~_E~BODIMENTS
Through the use of recombinant DNA methods, the
present inventors have identified novel mammalian receptor-
type (transmembrane) protein tyrosine phosphatases (PTPase;
EC 3.1.3.48). The murine R-PTPase-~ has 794 amino acids,
30 whereas the human R-PTPase-~ has 802 ~mino acids. In view
of its receptor-like structure, and t:he likelihood tha~ it
is part of a family, the inventors have termed this protein,
R-PTPase-~ (Eeceptor ~rotein tvrosine ~hosphatase alpha).
The family is designated herein as thll~ "R-PTPases."




.; . . ~ :

- . . ~ ~.

~140 92/010~;0 ~ /u~ sy~
2(~ 70~8

- 13 -
R-PTPase-~ has an intracellular domain homologous
to the catalytic domains of other tyrosine phosphatases.
The inventors have further characteri.zed the 142 amino acid
extracellular domain (including signal peptide) as having a
5 high serine and threonine content (32t) and 8 potential N-
glycosylation sites. The inventors have produced cDNA
clones coding for the novel protein, and expressed the
protein from eukaryotic hosts. Northern analysis has been
used to identify the natural expression of the protein in
10 various cells and tissues. They have further produced a
polyclonal antibody to the protein by immunization with a
synthetic peptide of R-PTPase-~, which identifies a 130 kDa
protein in cells transfected with a cDNA clone encoding a
portion of R-PTPase-~.
~emarkably, in addition to being composed of
intracellular domains having enzymatic activity, the
receptor family to which R-PTPases belong includes
transmembrane proteins having and N-terminal extracellular
domains; this is analogous to the tyrosine kinase enzyme
20 family (Tonks, N.K. et al. (1988) Bioche~istry 27, 8695-
8701; Charbonneau, H. et al. (1988) ~oc. Natl. Acad. Sci.
USA 85, 7182-7186; Streuli, M. et al., (1988) J. Exp. Med.
168, 1523- 2530; Streuli, M. et al., ~1989) Proc. Natl.
Acad. Sci. USA 86, 8698-8702). The present inventors have
25 therefore concluded that ligands in the extracellular
environment can control the activity of this membrane-
associated subclass of PTPases.
R-PTPase-~ and the other R-PTPases of the present
invention are useful in methods for sc:reening drugs and
30 other agents which are capable of activating or inhibiting
the R-PTPase activity, and thereby affecting major pathways
of cellular metabolism. 8y attaching an intact R-PTPase, or
the ligand-binding portion thereof, to a solid phase matrix,
an affinity probe is creat~d which can be used to screen
3' bioIogical products or chemical agents' for their capacity to




. . ., , ,, . . .i , ~ . , . ~.. -

~ L/ U I V:~U r~l/uaYI/uqnYL
2l~7~a8


interact with the receptor on the basis of their bindinq
activity. ~ound material can then be eluted from the
affinity probe in purified form.
Methods for coupling proteins and peptides to the
S solid phase, the solid phase substa~ces useful in these
methods, and means for elution, are well known to those of
skill in the art.
The R-PTPase protein or derivatives thereof having
enzymatic activity can be used for testing of compounds
10 capable of enhancing or inhibiting the phosphatase activity.
The ability of a compound under test t~ modify phosphatase
activity can be tested in an in vitro system wherein the
test compound is added to purified R-PTPase protein or
enzymatically active derivatives thereof, and the affects on
15 enzyme activity measured using standard enzymological
procedures well known to those of skill in the art.
Alternatively, the action of a compound on R-
PTPase activity can be measured in a whole cell preparation
using live or fixed cells, or a membrane fraction derived
20 from live or fixed cells. This method is useful for
screening compounds acting via the extracellular receptor
portion of the protein, as well as compounds acting directly
on the enzymatic portion of the protein. A test compound is
incubated with cells, or with a membrane preparation
25 derived therefrom, which express high amounts of the R- -
PTPase of this invention, such as transfected COS or NIH-3T3
cells. The amount of cellular phosphotyrosine is then
measured, using methods well-known in the art (Honegger,
A.M. et al~, Cell ~:199-209 (1987); Margolis, B. et al.,
30 Cell 5?:1101-1107 (1989)). The results are compared to
results obtained in the absence of the test compound, or in
the absence or presence of a known activator of R-PTPase.
~n such studies, the action of the test compound in the
presence of an activator of tyrosine kinase can also be
3~ measured.




- . . ................... ..
.,~ .

20~ 7~8

- 15 -
A compound which stimulates R-PTPase activity will
result in a ne~ decrease in the amount of phosphotyrosine,
whereas a compound which inhibits R-PTPase activity will
result in a net increase in the amount of phosphotyrosine.
In the case of growth factor receptors which are
tyrosine kinases, such as the receptors for epidermal growth
factor (EGF) and for platelet-derived growth factor (PDGF),
tyrosine phosphorylation is linked to cell growth and to
oncogenic transformation. Activation of a PTPases, leading
10 to dephosphorylation, would serve as a ~ounterregulatory
mechanism to prevent cr inhibit growth, and might serve as
an endogenous regulatory mechanism ag~inst cancer. Thus,
mutation or dysregulation of this receptor/enzyme system may
promote susceptibility to cancer
The insulin receptor is also a tyrosine kinase,-
and phosphorylation of tyrosine in cells bearing insulin
receptors would be associated with normal physiological
function. In contrast to the case of cell growth and
cancer, activation of an R-PTPase would counteract insulin
20 effects. Subnormal R-PTPase levels or enzymatic activity
would act to remove a normal counterregulatory mechanisms.
Perhaps more important, though, over-activity, or
inappropriate activation, of a R-PTPase would be expected to
inhibit or totally prevent the action of insulin on cells,
25 leading to diabetes (of an insulin-resistant variety).
Thus, susceptibility to diabetes may be associated with R-
PTPase dysregulation. ~ ;
Therefore, the methods of the present invention -~
for identifying normal or mutant R-PTPase genes, or for
30 measuring the amount or activity of R-PTPase associated with
a cell or tissue, can serve as method~ for identifying .
susceptibility to cancer, diabetes, or other diseases
associated with alterations in cellular phosphotyrosine
metabolism.

~-~ 7~U~V~V r~IIuaYI~ o~L
200 7~)3~

- 16 -
The present invention provides methods for
evaluating the presence and the level of normal or mutant R-
PTPase in a subject. Absence, or more typically, low
expression of the R-PTPase, or presence of a mutant R-
5 PTPase, in an individual may serve as an important predictorof susceptibility to oncogenic transformation and the
development of cancer. Alternatively, over-expression of R-
PTPase, possibly due to a mutant receptor/enzyme system
insensitive to negative regulation, or due to overabundance
10 of a stimulatory ligand in the body, may serve as an
important predictor of susceptibility to diabetes.
Oligonucleotide probes encoding various portions
of the R-PTPase (see below) are used to test cells from a
subject for the presence DNA or RNA sequences encoding the
15 R-PTPase. A preferred probe would be one directed to the
nucleic acid sequence encoding at least 4 amino acid
residues, and preferably at least 5 amino acid residues, of
the R-PTPase-~ or other R-PTPase protein of the present
invention. Qualitative or quantitative assays can be
20 performed using such probes. For example, Northern analysis
(see Examples III and VI, below) is used to measure
expression of an R-PTPase mRNA in a cell or tissue
preparation.
Such methods can be used even with very small
25 amounts of DNA obtained from an individual, following use of
selective amplification techniques. Recombinant DNA
methodologies capable of hmplifying purified nucleic acid
fragments have long been recognized. Typically, such
methodologies involve the introduction of the nucleic acid
30 fragment into a DNA or RNA vector, the clonal amplification
of the vector, and the recovery of the amplified nucleic
acid fragment. Examples of such methodologies are pro--ided
by Cohen et al. (U.S. Patent 4,237,224), SambrooX et al.
Nolecylar Clonin8: Al~aboratory Manual, Second Edition, Cold




: ., :. : ~, . , . :
..: , , ,: . . . . -.: : : : , ,
,

... .

2~o 7~

- 17 -
Spring Harbor Press, Cold Spring Harl~or, NY (1989), which
references are herein incorporated by reference).
Recently, an in vitro/ enz~matic method has been
described which is capable of incre2sing the concentration
5 of such desired nucleic acid molecules. This method has been
referred to as the "polymerase chain reaction or "PCR"
(Mullis, K. et al., Cold Sprina HarbQ Symp. Ouant. ~iol.
S1:263-273 (1986); Erlich, H. ~_g~, EP 50,424; EP
84,796, EP 258,017, EP 237,362; Mulli.s, K., EP 201,184;
lo Mullis, K. et al., US 4,683,202; Erlich, H., US 4,582,788;
and Saiki, R. et al., Us 4,683,194).
The polymerase chain reaction provides a method
for selectively increasing the concentration of a particular
nucleic acid sequence even when that sequence has not been
15 previously purified and is present only in a single copy in
a particular sample. The method can be used to amplify
either single- or double-stranded DNA. The essence of the
method involves the use of two oligonucleotide pro~es to
serve as primers for the template-dependent, polymerase
20 mediated replication of a desired nucleic acid molecule.
The precise nature of the two oligonucleotide
probes of the PCR method is critical to the success of the
method. As is well known, a molecule of DNA or RNA
possesses directionality, which is conferred through the 5'-
25 3' linkage of the phosphate groups of the molecule.Sequences of DNA or RNA are linked together through the
formation of a phosphodiester bond between the terminal 5'
phosphate group of one sequence and the terminal 3' hydroxyl
group of a second sequence. Polymerase dependent
30 amplification of a nucleic acid molecule proceeds by the
addition of a 5' nucleotide triphosphate to the 3' hydroxyl
end of a nucleic acid molecule. Thus~ the action of a
polymerase extends the 3' end of a nucleic acid molecule.
These inherent properties are exploited in the selection of
3~ the oligonucleotide probes of the PCR. The oligonucleotide




... , .... . - . . . .................. . , . :~


.

V ~ V~7~ ~ V_~17~
2U~ 7~08

- 18 -
sequences of the probes of the PCR mel:hod are selected such
that they contain sequences identical to, or complementary
to, sequences which flank the particular nucleic acid
sequence whose amplification is desire!d.
More specifically, the oligonucleotide sequences
of the "first" probe is selected such that it is capable of
hy~ridizing to an oligonucleotide sequence located 3~ to the
desired sequence, whereas the oligonuc:leotide sequence of
the "second" probe is selected such that it contains an
1~ oligonucleotide sequence identical to one present 5' to the
desired region. Both probes possess 3' hydroxy groups, and
therefore can serve as primers for nut:leic acid synthesis.
In the PCR, the reaction conditions are cycled
between those conducive to hybridization and nucleic acid
15 polymerization, and those which result in the denaturation
of duplex molecules. In the first st;ep of the reaction, the
nucleic acids of the sample are transiently heated, and then
cooled, in order to denature any double-stranded molecules
which may be present. The "first" and "second" probes are
20 then added to the sample at a concentxation which greatly
exceeds that of the desired nucleic acoid molecule. ~hen the
sample is incubated under conditions conducive to
hybridization and polymerization, the "first" probe will
hybridize to the nucleic acid molecule of the sample at a
25 position 3' to the sequence to be amplified. If the nucleic
acid molecule of the sample was initially double-stranded,
the "second" probe will hybridize to the complementary
strand of the nucleic acid ~olecule at a position 3' to the
sequence which is the complement of the sequence whose
30 amplification is desired. Upon addition of a polymerase,
the 3' ends of the "fir~t" and (if the nucleic acid molecule
was double-stranded) "second" probes will be extended. The
extension of the "first" pr~be will re~sult in the synthesis
of an oligonucleotide having the exact seguence of the
35 desired nucleic acid. Extension of the "second" probe will




:, . .. .
: : : :: .

... .. . .. , . . - .. . . :. .. . . . ..
. .
. : ~ :. .
. .

'1 / U~ 1 /U4~9Z
2~7008

-- 19 --
result in the synthesis of an oligonucleotide having the
exact sequence of the complement of t:he desired nucleic
acid.
The PCR reaction is capable of exponential
5 amplification of specific nucleic aci.d sequences because the
extension product of the "first" proble, of necessity,
contains a sequence which is complementary to a sequence of
the "second" probe, and thus can serve as a template for the
production of an extension product of the "second" probe.
10 Similarly, the extension product of the "second" probe, of
necessity, contains a sequence which is complementary to a
seguence of the "first" probe, and th~s can serve as a
template for the production of an extension product of the
"first" probe. Thus, by permitting cycles of
15 polymerization, and denaturation, a geometric increase in
the concentration of the desired nucleic acid molecule can
be achieved. Reviews of the PCR are provided by Mullis,
K-B- (Col_ sp~ins~ kQr Sy~r. oua~ Biol. ~:263-273
(1986)); Saiki, R.X., çt al. (Bio/~echnLology 3:1008-1012
~^ (1985)); and Mullis, R.B., et_al. (Meth. ~n~Y~ol. 155:335_
350 (1987)).
In one embodiment, the invention i5 directed to a
naturally occurring mammalian R-PTPase~. In another
embodiment, the invention is directed to a recombinant
25 mammalian R-PTPase-~. The preferred R-P~Pases of the
present invention are of human origin. The invention
provides the naturally occurring molecule substantially free
of other proteins with which it is natively associated. ~-
~Substantially free of other proteins or glycoproteins"
30 indicates that the protein has been purified away from at
least 90 per cent (on a weight basis) and from even at
least 99 per cent if desired, of other proteins and
glycoproteins with which it is nati-vely associated, and is
therefore substantially free of them. That can be achieved
35 by subjecting the cells, tissue or fluids containing the R-


~..




' , . . ' ~ ' ' ' ' ' ., .

) 0 8

- 20 -
PTPase to standard protein purification techniques such as
immunoadsorbent columns bearing monoclonal antibodies
reactive against the protein. Other ~orms of affinity
purification can utilize solid-phase substrates which can
5 bind the PTPase domain, or a ligand t:hat will bind to the
receptor domain. Alternatively, the purification can be
achieved by a combination of standarcl methods, such as
ammonium sulfate precipitation, molec:ular sieve
chromatography, and ion exchange chromatography.
It will be understood that t:he mammalian R-PTPase
of the present invention can be bioch~mically purified from
a variety of cell or tissue sources. For preparation of
naturally occurring R-PTPase, tissues; such as mammalian
placenta or brain, especially of human origin, are
15 preferred.
Alternatively, because the gene for the R-PTPase
can be isolated or synthesized, the polypeptide can be
synthesized substantially free of other proteins or
glycoproteins of mammalian origin in a prokaryotic organism
20 or in a non-mammalian eukaryotic organism, if desired. As
intended by the present invention, a recombinant R-PTPase-
~molecule produced in mammalian cells, such as transfected
COS, NIH-3T3, or CHO cells, for sxamE~le, is either a
naturally occurring protein sequence or a functional
25 derivative thereof. Where a naturally occurring protein or
glycoprotein is produced by recombinant means, it is
provided substantially free of the ot:h~er prot~ins and
glycoproteins with which it is natively associated.
Alternatively, methods are w~ell known for the
30 synthesis of polypeptides of de~ired ~equence on solid phase
supports and their 6ubsequent separat:i.on from the support.
In a further embodiment, the invention provides
"functional derivatives" of the R-PTPase~ ~y "functional
derivative" is meant a "fragment," "variant," "analog," or




~, . .. , .. . .... : ,
.- : . .... . .,, .-

U I V J ~ V ~
2~ )0~

- 21 -
~chemical derivative~ of the R-PTPasle, which terms are
defined below. A function
al derivative retains at least a portion of the function of
the R-PTPase, such as binding to a specific antibody,
5 phosphatase enzymatic activity or binding of the
extracellular domain to a ligand, which permits its utility
in accordance with the present invention.
A "fragment" of the R-P~Pase re~ers to any subset
of the molecule, that is, a shorter peptide.
13 A "variant" of the R-PTPase refers to a molecule
substantially similar to either the entire peptide or a
fragment thereof. Variant peptides may be conveniently
prepared by direct chemical synthesis of the variant
peptide, using methods well- known in the art.
Alternatively, amino acid s;equence variants of the
peptide can be prepared by mutations in the DNA which
encodes the synthesized peptide. Such variants include, for
example, deletions from, or insertions or substitutions of,
residues within the amino acid sequence. Any combination of
20 deletion, insertion, and substitution may ~lso be made to
arrive at the final construct, providled that the final
construct possesses the desired activity. Obviously, the
mutations that will be made in the DNA encoding the variant
peptide must not alter the reading frame and preferably will
25 not create complementary regions that could produce
secondary mRNA structure ~see European Patent Publication
No. EP 75,444).
At the genetic level, these variants ordinarily
are prepared by site-directed mutagenesis (as exempl$fied by
30 Adelman et al., ~ 2:183 (1983)) of nucleotides in the DNA
encoding the peptide molecule, thereby producing DNA
encoding the variant, and thereafter expressing the DNA in
recombinant cell culture (see below). The variants
typically exhibit the same qualitative biological activity
3 r as the nonvariant peptide.




.. .. . . .. , . . ,. . . , . ~ : :

:. ... ~

. .
, , ~ .. . . .. . .

2~70~8

- 22 -
An ~analog~ of the R-PTPase refers to a non-
natural molecule substantially similar to either the entire
molecule or a fragment t~ereof.
A "chemical derivative" of the R-PTPase contains
5 additional chemical moieties not normally a part of the
peptide. Covalent modifications of the peptide are included
within the scope of this invention. Such modifications may
be introduced into the molecule by reacting targeted amino
acid residues of the peptide with an organic deri~atizing
10 agent that is capable of reacting with selected side chains
or terminal residues.
Cysteinyl residues most commonly are reacted with
alpha-haloacetates (and corresponding amines), such as
chloroacetic acid or chloroacetamide, to give carboxymethyl
lS or carboxyamidomethyl derivatives. Cysteinyl residues also
are derivatized by reaction with bromotrifluoroacetone,
alpha-bromo- beta-(5-i~idozoyl)propionic acid, chloroacetyl
phosphate, N- alkylmaleimides, 3-nitro-2-pyridyl disulfide,
methyl 2-pyridyl disulfide, p-chloromercuribenzoate, 2-
20 chloromercuri-4- nitrophenol, or chloro-7-nitrobenzo-2-oxa-
1,3-diazole.
Histidyl residues are derivatized by reaction with
diethylprocarbonate at pH 5.5-7.0 because this agent is
relatively specific for the histidyl side chain. Para-
25 bromophenacyl bromide also is useful; the reaction ispreferably performed in 0.1 M sodium cacodylate at pH 6Ø
Lysinyl and amino termina; residues ~re reacted
with succinic or other carboxylic acid anhydrides.
Derivatization with these agents has the effect of reversing
30 the charge of the lysinyl residues. Other suitable reagents
for derivatizing alpha-amino-containing residues include
imidoesters such as methyl picolinimidate; pyridoxal
phosphate; pyridoxal; chloroborohydride;
trinitrobenzenesulfonic acid; O-methylisourea; 2,4




" : . ' ., ' ': .: ' . .' . ' ' ' ' ' ' ' ' '; ' ' i ' ~ ' : : .

:: ' .' ' ' ' ~ - :." : , ' ': -, ' ,

I V ~ V J ~
20~703~

- 23 -
pentanedione; and transaminase-catalyzed reaction with
glyoxylate.
Arginyl residues are modified by reaction with one
or ~everal conventional reagents, among them phen~lglyoxal,
5 2,3- butanedione, 1,2-cyclohexanedione, and ninhydrin.
Derivatization of arginine residues re~uires that the
reaction be performed in alkaline conditions because of the
high PKa of the guanidine functional group. Further~ore,
these reagents may react with the groups of lysine as well
lC as the arginine epsilon-amino group.
The specific modification of tyrosyl residues ~E
se has been studied extensively, with particular interest in
introducing spectral labels into tyrosyl residues by
reaction with aromatic diazonium compounds or
15 tetranitromethane. Most commonly, N-acetylimidizol and
tetranitromethane are used to form O-acetyl tyrosyl species
and 3-nitro derivatives, respectively.
Carboxyl side groups (aspartyl or glutamyl) are
selectively modified by reaction with carbodiimides (R'-N-C-
20 N-R') such as l-cyclohexy1-3-(2-morpholinyl-(4-ethyl)
carbodiimide or 1- ethyl-3-(4-azonia-4,4-dimethylpentyl)
carbodiimide. Furthermore, aspartyl and glutamyl residues
are converted to asparaginyl and glutaminyl residues by
reaction with ammonium ions.
Glutaminyl and asparaginyl residues are frequently
deamidated to the corresponding glutamyl and aspartyl
residues. Alternatively, these residues are deamidated
under mildly acidic conditions. Either for~ of these
residues falls within the scope of this invention.
Derivatization with bifunctional agents is useful
for cross-linking the peptide to a wa1;er-insoluble support
matrix or to other macromolecular carriers. Commonly used
cross-linking agents include, e.g., 1,1-bis(diazoacetyl)-2-
phenylethane, glutaraldehyde, N-hydroxysuccinimide esters,
35 for example, esters with 4-azidosalicylic acid,




, . :. : :- : - - : . .:

.. . . . . . .. . . . . ..

20~7038

- 24 -
homobifunctional imidoesters, including disuccinimidyl
esters such as 3,3~- dithiobis~sucoinimidyl~propionate), and
bifunctional maleimides such as bis-N-maleimido-1,8-octane.
Derivatizing agents such as methyl-3-[tp-
5 azidophenyl)dithio]propioimidate yield photoactivatableintermediates that are capable of forming crosslinks in the
presence of light. Alternatively, reactive water-insoluble
matrices such as cyanogen bromide-activated carbo
hydrates and the reactive substrates described in U.S.
10 Patent Nos. 3,969,287+ 3,691,016; 4,195,128; 4,247,642;
4,229,537; and 4,330,440 are employed for protein
immobilization.
Other modifications include hydroxylation of
proline and lysine, phosphorylation of hydroxyl groups of
15 seryl or thrPo~yl residues, methylation of the alpha-amino
groups of lysine, arginine, and histidine side chains (T.E.
Creighton, Proteins: Structure and Molecule Properties,
W.H. Freeman & Co., San Francisco, pp. 79-86 (1983)),
acetylation of the N-terminal amine, and, in some instances,
20 amidation of the C-terminal carboxyl groups.
Such derivatized moieties may improve the
solubility, absorption, biological half life, and the like.
The moieties may alternatively eliminate or attenuate any
undesirable side effect of the protein and the like.
2S Moieties capable of mediating such effects are disclosed,
for example, in ~emin~ton's Pharmaceutical Sciences, 16th
ed., Mack Publishing Co., Easton, PA ~1980)
This invention is also directed to an antibody
specific for an epitope of R-PTPase, preferably, of R-
30 PTPase-~, most preferably of human R-PTPase-~, and the use
of such antibody to detect the presenoe of, or measure the
quantity or concentration of, the R-PY'Pase in a cell, a cell
or tissue extract, or a biological fluid.
The term "antibody" is meant to include polyclonal
.




. . .: . . . .
;. ~ - . , . .. :, :
,,: -

.
. i - . . . . - .: . : ~ . ~-... . - , -

20~:~7038


antibodies, monoclonal antibodies (mAbs), chimeric "
antibodies, and anti-idiotypic (anti-Id) antibodies.
Polyclonal antibodies are heterogeneous
populations of antibody molecules derived from the sera of
5 animals immunized with an antigen.
Monoclonal antibodies are a substantially
~ homogeneous population of antibodies to spe~ific antigens.
- MAbs may be obtained by methods known to those skilled in
the art. See, for example Kohler and Milstein, Nature
10 256:495-497 (1975) and U.S. Patent No. 4,376,110. Such
antibodies may be of any immunoglobulin class including IgG,
IgM, IgE, IgA, GILD and any subclass thereof. The
hybridoma producing the mAbs of this invention may be
cultivated in vitro or in vivo. Production of high titers
15 of mAbs in vivo production makes this the presently
preferred method of production. Brie~ly, cells from the
individual hybridomas are injected intraperitoneally into
pristane-primed BAL~/c mice to produce ascites fluid
containing high concentrations of the desired mAbs. MAbs of
20 isotype IgM or IgG may be purified from such ascites fluids,
; or from culture supernatants, using column chromatography
methods well known to those of skill in the axt.
Chimeric antibo~ies are molecules different
portions of which are derived from diferent animal species
25 , such as those having variable region derived from a murine
mAb and a human immunoglobulin constant region. Chimeric
antibodies and methods for their production are known in the
art ( Cabilly et al, Proc. Natl. Acad. S~i. U~ 3273-
3277 (1984); Morrison et al!, Proc. Natl. Acad. Sci. USA
30 81:6851-6855 (1984); Boulianne et al., Nature 312:643-646
(1984); Cabilly et al., European Patent Application 125023
(published November 14, 1984); Neuberger et al., Nature
! 314:268-270 (1985); Taniguchi et al., European Patent
Application 171496 (published February 19, 1985); Morrison
3~ et al., European Patent Application 173494 (published March

2~ 7~8

- 26 -
5, 1986~; Neuberger et al., PCT Application W0 86/01533
(published March 13, 1986); Kudo et al., European Patent
Application 184187 (published June 11, 1986); Morrison
et al., European Patent Application 173494 (published March
5 5, 1986); Sahagan et al., J. Immuno,L 137:1066-1074 (1986);
Robinson et al., International Patent Publication
#PCT/US86/02269 (published 7 May 1987); Liu et al., Proc.
Natl. Acad. Sci. USA 84:3439-3443 (:Ls87); Sun et ~l., Proc.
Natl. Acad. Sci. USA ~4:214-218 (1987~; Better et al.,
10 Scienc 240:1041- 1043 (1988)). Theæe references are hereby
incorporated by reference.
An anti-idiotypic (anti-Id) antibody is an
antibody which recognizes unique det:erminants generally
associated with the antigen-binding site of an antibody. An
15 anti-Id antibody can be prepared by i~munizing an animal of
the same species and genetic type (e.g. mouse strain) as the
source of the mAb with the mAb to which an anti-Id is being
prepared. The immunized animal will recognize and respond
to the idiotypic determinants of the immunizing antibody by
20 producing an antibody to these idiotypic determinants (the
anti-Id antibody).
The anti-Id antibody may also be used as an
"immunogen" to induce an immune respo~se in yet another
animal, producing a so-called anti-anti-Id antibody. The
25 anti-anti-Id may be epitopically identical to the original
mAb which induced the anti-Id. ~hus, by using antibodies to
the idiotypic determinants of a mAb, it is possible to
identify other clones expressing antibodies of identical
specificity.
Accordingly, mAbs generated against the R-PTPase
of the present invention may be used to induce anti-Id
antibodies in suitable animals, such 8S BALB/c mice. Spleen
cells from such immunized mice are used to produce anti-Id
hybridomas secreting anti-Id mAb8. Further, the anti-Id .
35 mAbs can be coupled to a carrier such as keyhole limpet

WO 9Z/U 1 t~5U r~, 1 / u~ 07L
7 ~ 0 8


hemocyanin (KLH) and used to immuniz6! additional BALB/c
mice. Sera from these mice will cont:c~lin anti-anti-Id
antibodies that have the binding prop~ rties of the original
mAb specific for a R-PTPase epitope.
The anti-Id mAbs thus have 1heir own idiotypic
epitopes, or ~idiotopes~ structurally similar to the epitope
being evaluated, such as R-PTPase-~.
The term "antibody" is alscl meant to include both
intact molecules as well as fragments thereof, such as, for
10 example, Fab and F(ab')2, which are capable of binding
antigen. Fab and F(ab'~ fragments icLck the Fc fragment of
intact antibody, clear more rapidly fIom the circulation,
and may have less non-specific tissue binding than an intact
antibody (Wahl et al., J. Nucl. Med. ~4:316-325 (1983)).
It will be appreciated that Fab and F(ab')2 and
other fragments of the antibodies uselul in the present
invention may be used for the detectia,n and quantitation of
R-PTPase according to the methods disc:losed herein for
intact antibody molecules. . Such f~agments are typically
~0 produced by proteolytic cleavage, using enzymes such as
papain (to produce Fab fragments) or pepsin (to produce
F(ab')2 fragments).
An antibody is said to be "c:apable of binding" a
molecule if it is capable of specifically reacting with the
25 molecule to thereby bind the molecule to the antibody. The
term "epitope" is meant to refer to that portion of any
molecule capable of being bound by an antibody which can
also be recognized by that antibody. Epitopes or "antigenic
determinants" usually consist of chemically active surface
30 groupings of molecules such as ~mino zcids or sugar side
chains and have specific three dimens:onal structural
characteristics as well as specific charge characteristics.
An ~'antigen" is a molecule or a portion of a
molecule capable of being bound by an antibody which is
35 additionally capable of inducing an ani~al to produce



, . - ~, . , . : .
.,, . . . . : ~. ,.
.
:

- ~: - ., . ~- : ,
:

WO92/010~0 PCT/US91~92
2~, 70 ~8

- 28 -
antibody capable of binding to an epitope of that antigen.
An antigen may have one, or more than one epitope. The
specific reaction referred to above ils meant to indicate
that the antigen will react, in a highly selective manner,
5 with its corresponding antibody and ;not with the multitude
of other antibodies which may be evoked by other antigens.
The antibodies, or fragmen~s of antibodies, useful
in the present invention may be used I:o ~uantitatively or
qualitatively detect the presence of c:ells which express the
10 R-PTPase protein. This can be accomplished by
immunofluorescence techniques employing a fluorescently
labeled antibody (see below) coupled with light microscopic,
flow cytometric, or fluorimetric detec:tion.
The antibodies (of fragments thereof) useful in
15 the present invention may be employed hictologically, as in
immunofluorescence or immunoelectron microscopy, for in situ
detection of R-PTPase. In ~i~ detec:tion may be
accomplished by removing a histologic:al specimen from a
patient, and providing the a labeled antibody of the present
20 invention to such a specimen. The antibody (or fragment) is
preferably provided by applying or by overlaying the labeled
antibody (or fragment) to a biological sample. Through the
use of such a procedure, it is possible to determine not
only the presence of the R-PTPase but: also its distribution
25 on the examined tissue. Using the present invention, those
of ordinary skill will readily percei.ve that any of a wide
variety of histological methods (such as staining
procedures) can be modified in order to achieve such in situ
detection. Such assays for R-PTPase typically comprises
30 incubating a biological sample, such as a biological fluid,
a tissue extract, freshly harvested cells such as
lymphocytes or leucocytes, or cells which have been
incubated in tissue culture, in the presence of a detectably
labeled antibody capable of identifying R-PTPase, and




` ' : .. '; . ' '. ` `' ' .'` , ' , : : `': . ' ~ .' ',,' . . ' : ` ' .`

~ V 7 ~ 1 V ~ ~J J V ~ v ~

2~)(,7~8

- 2g -
detecting the antibody ~y any of a number of techniques
well-known in the art.
The biological sample may be treated with a solid
phase support such as nitrocellulose, or other solid support
5 which is capable of immobilizing cells, cell particles or
soluble proteins. The support may then be washed with
suitable buffers followed by treatment with the detectably
labeled R-PTPase-specific antibody. The solid phase support
may then be washed with the buffer a second time to remove
10 unbound antibody. The amount of bound label on said solid
support may then be detected by conventional means.
By "solid phase support" is intended any support
capable of binding antigen or antibodies. Well-known
supports, or carriers, include glass, polystyrene,
15 polypropylene, polyethylene, dextran, nylon, amylases,
natural and modified celluloses, polyacrylamides, gabbros,
and magnetite. The nature of the carrier can be either
soluble to some extent or insoluble for the purposes of the
present invention. The support material may have virtually
20 any possible structural configuration so long as the coupled
molecule is capable of binding to an antigen or antibody.
Thus, the support configuration may be spherical, as in a
bead, or cylindrical, as in the inside surface of a test
tube, or the external surface of a rod. Alternatively, the
25 surface may be flat such as a sheet, test strip, etc.
Preferred supports include polystyrene beads. Those sXilled
in the art will know many other suitable carriers for
binding antibody or antigen, or will be able to ascertain
the same by use of routine experimentation.
The binding activity of a givén lot of anti-R-
PTPase antibody may be determined according to well known
methods. Those skilled in the art will be able to determine
operative and optimal assay conditions for each
determination by employing routine experimentation.




' ~ ~ ; : .. . . .

~J Y'/UII)~I) PCl/US91/0489Z
2~ ~ 0~8

- 30 -
Other such steps as washing, stirring, shaking,
filtering and the like may be added to the assays as is
customary or necessary for the particular situation.
One of the ways in which the R-PTPase-specific
5 antibody ca~ be detectably labeled is by linking the same to
an enzyme and use in an enzyme immunoassay (EIA). This
enzyme, in turn, when later exposed to an appropriate
substrate, will react with the substrate in such a ~anner as
to produce a chemical moiety which can be detected, for
10 example, by spectrophotometric, fluorimetric or by visual
means. Enzymes which can be used to detectably label the
antibody include, but are not limited to, malate
dehydrogenase, staphylococcal nuclease, delta-5-steroid
isomerase, yeast alcohol dehydrogenase, alpha-
15 glycerophosphate dehydrogenase, triose phosphate isomerase,horseradish peroxidase, alkaline phosphatase, asparaginase,
glucose oxidase, beta-galactosidase, ribonuclease, urease,
catalase, glucose-6-phosphate dehydrogenase, glucoamylase
and acetylcholinesterase. The detection can be accomplished
20 by colorimetric methods which employ a chro~ogenic substrate
for the enzyme. Detection may aiso be accomplished by
visual comparison of the extent of enzymatic reaction of a
substrate in comparison with similarly prepared standards.
Detection may be accomplished using any of a
25 variety of other immunoassays. For example, by
radioactively l~beling the antibodies or antibody fragments,
lt is possible to detect R-PTPase through the use of a
radioimmunoassay (RIA) (see, for example, Work, T.S. et al.,
Labo~atory Techniques and Biochemist~ in ~Q~ecu~Lar ~ioloqy,
3D North Holland Publishing Company, New York, 1978, which is
$ncorporated by reference herein). The radioactive isotope
can be detected by such means as the use of a gamma counter
or a scintillation counter or by autoradiography.
It is also possible to label the antibody with a
35 fluorescent compound. When the fluorescently labeled ~ -
,




.. . . ..

" u: u r-_'1/ IJ3~ 1~U'~
2l3o7008


antibody is exposed to light of the proper wave length, its
presence can then be detectecl due to fluorescence. Among
the most commonly used fluorescent labelling compounds are
fluorescein isothiocyanate, rhodamine, phycoerythrin,
5 phycocyanin, allophycocyanin, Q- phthaldehyde and
fluorescami~e.
The antibody can also be deteotably labeled using
fluoresoence emitting metals such as 152Eu, or others of the
lanthanide series. These metals can be attached to the
10 antibody using such metal chelating group~ as
diethylenetriaminepentaacetic acid (DTPA) or
ethylenediaminetetraacetic acid ~ED~A).
The anti~ody also can be detectably labeled by
coupling it to a chemiluminescent compound. The presence of
15 the chemiluminescent-tagged antibody is then determined by
detecting the presence of luminescence that arises during
the course of a chemical reaction. Examples of particularly
useful chemiluminescent labeling compounds are luminol,
isoluminol, theromatic acridinium ester, imidazole,
20 acridinium salt and oxalate ester.
Likewise, a bioluminescent compound may be used to
label the antibody of the present invention.
Bioluminescence is a type of chemiluminescence found in
biological systems in which a catalytic protein increases
2S the efficiency of the chemiluminescent reaction. The
presence of a ~ioiuminescent protein is determined ~y
detecting the presence of luminescence. Important
bioluminescent compounds for purposes of labeling are
luciferin, luciferase and aeguorin.
The antibody molecules of the present invention
may be adapted for utilization in an :mmunometric assay,
also known as a "two-site" or "sandwich" assay. In a
typical immunometric assay, a quantity of unlabeled antibody
~or fragment of antibody) is bound to a solid support and a
35 quantity of detectably labeled soluble antibody is added to




- - , . . .



, , :
: . . . . . .. . . .

2 ~ 0 8

- 32 -
permit detection and/or quantitation of the ternary complex
formed between solid-phase antibody~ antigen, and labeled
antibody.
Typical, and preferred, i~nunometric assays
5 include ~'forward~ assays in which the antibody bound to the
solid phase is first contacted with ~he sample being tested
to extract the antigen from the sample by formation of a
binary solid phase antibody-antigen c:omplex. After a
suitable incubation period, the solicl upport is washed to
10 remove the residue of the fluid sampl:l.e, including unreacted
antigen, if any, and then contacted with the solution
containing an unknown quantity of labeled antibody (which
functions as a "reporter molecule"). After a second
incubation period to permit the labeled antibody t~ complex
15 with the antigen bound to the solid H~upport through the
unlabeled antibody, the solid support is washed a second
time to remove the unreacted labeled antibody.
In another type of "sandwi~h" assay, which may
also be useful with the antigens of t:he present invention,
20 the so-called "simultaneous" and "re~erse" assays are used.
A simultaneous assay involves a single incubation step as
the antibody bound to the solid support and labeled antibody
are both added to the sample being tested at the same time.
After the incubation is completed, the solid support is
25 washed to remo~e the residue of fluid sample and uncomplexed
labeled antibody. The presence of labeled antibody
associated with the solid support is then determined as it
would be in a conventional "forward" sandwich assay.
In the "reverse" assay, ~tepwise addition first of
30 a solution of labeled antibody to the fluid sample followed
by the addition of unlabeled antibody bound to a ~olid
support after a suitable incubation period is utilized -
After a second incubation, the solid phase is washed in
conventional fashion to free it of thls residue of the-sample
35 being tested and the solution of unre!acted labeled antibody.




.. ...... . - . : : ' '' ' ' ' ' ~ ~ :' "' '' "' '

-. . . -: ' ' . : . :'
~ - . . . . .
: . . - - . ,.:,, -, ~ ., , : ~ .
. . ... . . . .
- . . : .: .. . - . : -
. : - : ::
: :.- ~ . :
. - ,

2~70~

- 33 -
The determination of labeled antibody associated with a
s~lid support is then determined as i~l the "simultaneousl'
and "forward~ assays.
The presence of normally fu~ctioning R-PTPase in a
5 subject can also be tested using direct enzymatic assays,
for the tyrosine phosphatase activity. Such biochemical
measurements can be performed in_vitro, using puri~ied
enzymes, allowing precise measurements ~f enzyme activity,
or with membrane preparations, or whole cells, where the net
10 phosphotyrosine level is determined.
In additional embodiments of the present
invention, a DNA sequence encoding a R-PTPase molecule and
methods for expressing the DNA sequence are provided. One
of ordinary skill in the art will know how to identify and
15 clone additional PTPase molecules, of human or other
mammalian species, which have seguence homology to the R-
PTPase molecules described herein, using the genetic
sequences and oligonucleotides of the present invention
without undue experimentation. Furthermore, manipulation of
20 the genetic constructs of the present invention allow the
grafting of a particular ligand-binding receptor domain onto
the transmembrane and catalytic portions of the R-PTPase
resulting in chimeric molecules. Non-limiting examples of
such chimeric molecules include the R~TPase wherein the
25 receptor is an epidermal growth factor receptor, a
fibroblast growth factor receptor, and the like.
Genetically engineered chimeric receptors are kn~wn in the
art (see, for example, Riedel, H. et ~1., Nature ~ 628-670
(1986)~.
Genetic constructs encoding R-PTPase-~, functional
derivative thereof, and chimeric molecules such ~s those
described above, can be used in gene therapy. An abnormal
or dysfunctional R-PTPase, which results in disease, may be
replaced by infusion of cells of the desired lineage (such
3' as hemopoietic cells, for example) transfected with a normal




. , ,, . . . - , .. , :
, . . , . . . ~ .... -, .. : ,,.-,. ., .. : . ., :... -
- , . - :
.. -.. , : :~ ., : , ,. :
-- , -.... ,:, :, . .. .
. , "; . .

. ,.

WO 92/01050 PCl'/US91/0489;!
2~)~700~

- 34 -
R-PTPase. Alternatively, or additioJIally, cells carrying a
chimeric R-PTPase having a receptor l:o a ligand of choice
(e.g. EGF) ca~ be used for such gene therapy.
The recombinant DNA molec~lles of the present
5 invention can be produced through any of a variety of means,
such as, for example, DNA or RNA synthesis, or more
preferably, by application of recombinant DNA techniques.
Techniques for synthesizing such molecules are disclosed ~y,
for example, Wu, R., et al. (Prog. ~lucl. Acid. Res. Molec.
10 Biol. 21:101-141 (1978)). Procedures for constructing
recombinant molecules in accordance with the above-
described method are disclosed by Sambrook et a L (supra).
The 3' terminus of the rec:ombinant molecule of
this $nvention is preferably treated to render it unsuitable
15 for polymerization. Such treatment may be accomplished by
blocking the terminus by chemical mealn6, or by modifying the
terminal bases such that they stericallly interfere with
polymerase action. In a preferred eDIbodiment, such
treatment is accomplishçd by immobilizing the 3' terminus,
20 such as by coupling it to a solid suE~port (~uch as, for
example, glass, plastic, latex, etc.). The support may be
of any form (i.e. a sheet, rod, sphere, ovoid, etc. Proce-
dures for such im~obilization are well known to those of
ordinary sXill. In the most preferred embodiment, the 3'
25 end of the recombinant molecule is covalently bound to the ~;~
solid support. A spacer region may be used to extend the
probe outward from the solid support as long as (1) it will
not sterically hinder any function or characteristic of the
recombinant molecule, and (2) the sequence of the spacer
30 region does not participate in the hybridization or
polymerization reactions of the assay. It is typically
desirable to immobilize several, and preferably, a large
number of such recombinant molecule to the ~upport.
Oligonucleotides representing a portion of an R-
35 PTPase are useful for screening for the presence of genes




.. . .:

2 ~ ~" J ~ 8

- 35 -
encoding such proteins and for the cloning of R-PTPase
genes. Techniques for synthesizing such oligonucleotides
are disclosed by, for example, Wu, R , et al., Pro~. Nucl.
Acid. Res. Molec. Biol. 21:101 141 (1978)).
Protein molecules are fragmented as with cyanogen
bromide, or with proteases such as papain, chymotrypsin,
trypsin, etc. ~Oike, Y., et al., J. ~iol. Chem. 257:9751-
9758 (1982); Liu, C., et al., Int. J, Pept. Protein Res.
21:209-215 (1983)). Because the genetic code is degenerate,
10 more than one codon may be used to encode a particular amino
acid (Watson, J.D., In: Molecular Bj,ology of the Gene, 4th
Ed., BenjamintCummings Publishing Co., Inc., Menlo Park, CA
(1987)). Using the genetic code, one! or more different
oligonucleotides can be identified, e!ach of which would be
15 capable of encoding the amino acid. The probability that a
particular oligonucleotide will, in iact, constitute the
actual XXX-encoding sequence can be estimated by considering
abnormal base pairing relationships and the frequency with
which a particular codon is actually used (to encode a
particular amino acid) in eukaryotic cells. Such "codon
usage rules" are disclosed by Lathe, R., et al., J. Molec.
Biol. 183:1-12 (1985). Using the "codon usage rules" of
Lathe, a single oligonucleotide, or a set of
oligonucleotides, that contains a theoretical "most
25 probable" nucleotide sequence capable of encoding the R-
PTPase sequences is identified.
Although occasionally an amino acid sequences may
be encoded by only a single oligonuc:l,eotide, frequently the
amino acid sequence may be encoded by any of a set of
30 similar oligonucleotides. Important:Ly, whereas all of the
members of this set contain oligonuc:Leotides which are
capable of encoding the peptide frag~ent and, thus,
potentially contain the same oligonucLeotide sequence as the
gene which encodes the peptide fragment, only one member of
35 the set contains the nucleotide sequence that is identical




....... : . , , - : .. . . -.. .:
- . -- - . ; : ,:
, ::, ~ - , , :.
. . ~ . . :. ~ , , ; .

, ~ ; :~, ,~ .. -... ... . .
., . .; .... .... . . .

2~ 17~8

- ~6 -
to the nucleotide sequence of the gene. Because this member
is present within the set, and is capable of hy~ridizing to
DNA even in the presence of the other me~bers of the set, it
is possible ~o employ the unfractionated set of
5 oligonucleotides in the same manner in which one would
employ a single oligonucleotide to clone the gene that
encodes the peptide.
The oligonucleotide, or set of oligonucleotides,
containing the theoretical "most pro~able" sequence capable
10 of encoding the R-PTPase fragment is used to identify the
; sequence of a complementary oligonucleotide or set of
oligonucleotides which is capable of hybridizing to the
"most probable" sequence, or set of sequences. An
oligonucleotide containing such a complementary sequence can
15 be employed as a probe to identify and isolate the R-PTPase
gene (Sambrook et al., supra).
A suitable oligonucleotide, or set of
oligonucleotides, which is capable of encoding a fragment of
the R-P~Pase gene (or which is complementary to such an
20 oligonucleotide, or set of oligonucleotides) is identified ~;
(using the above-described procedure), synthesized, and
hybridized by means well known in the art, against a DNA or, -~
more preferably, a cDNA preparation derived from cells which
are capable of expressing the R-PTPase gene. Single
25 stranded oligonucleotide molecules complementary to the
"most proba~le" R-PTPase peptide encoding sequences can be
synthesized using procedures which are well known to those
of ordinary skill in the art (Belagaje, R., et al., J. ~ol.
Chem. 254:5765-5780 (1979); Maniatis, T., et al., In:
30 Molecular Mechanisms in the Control of Gene Expression,
Nierlich, D.P., et ~1., Eds., Acad. Press, NY (1976); Wu,
R., çt al., Prw. Nucl. Acid Res. Molec. Biol. ~:101-141
(1978); Khorana, R.G., Science 203:614-625 (1979)).
Additionally, DNA synthesis may be achieved through the use
35 of automated synthesizers. Techniques of nucleic acid




......... ... ..... .. ... .... . .


, : - . : . - : : , ,. .: : : , : ~ - : . : : - .,, : , .: . :, . : . :

: - . . .
.: . .

~--v ~ ~ ~ u ~ v ~ v
2~(~70~8

- 37 -
hybridization are disclosed by Sambrook et al. (supra), and
by Haymes, B.D., et aL (In: Nucleic Acid Hybridization A
Practical Approach, IRL Press, Washington, DC (1985)), which
references are herein incorporated by reference. Techniques
5 such as, or similar to, those described above have
successfully enabled the cloning of genes for human aldehyde
dehydrogenases (Hsu, L.C., et al., Proc. Natl. Acad. Sci.
USA 82:3771-3775 (1985)), fibronectin (Suzuki, S., et al.,
EMBO J. 4:2519-2524 (1985)), the human estrogen receptor
lC gene (Walter, P., et al., Proc. Natl. Acad. Sci. USA
82:7889-7893 (1985)), tissue-type plasminogen activator
(Pennica, D., et al., Nature 301:214-221 (1983)) and human
term placental alkaline phosphatase complementary DNA tKam,
W., et al., Proc. Natl. Acad. Sci. USA 82:(715-8719
15 (1985)).
In a alternative way of cloning the R-PTPase gene,
a library of expression vectors is prepared by cloning DNA
or, more preferably, cDNA (from a cell capable of expressing
R-PTPase) into an expression vector. The library is then
20 screened for members capable of expressing a protein which
binds to anti-R-PTPase antibody, and which has a nucleotide
sequence that is capable of encoding polypeptides that have
the same amino acid sequence as R-PTPase, or fragments
thereof. In this embodiment, DNA, or more preferably cDNA,
25 is extracted and purified from a cell which is capable of
expressing R-PTPase protein. The purified cDNA is
fragmented (by shearing, endonuclease digestion, etc.) to
produce a pool of DNA or cDNA fragments. DNA or cDNA
fragments from this pool are then cloned into an expression
30 vector in order to produce a genomic library of expression
vectors whose members each contain a unique cloned DNA or
cDNA fragment.
An "expression vector" is a vector which (due to
the presence of appropriate transcriptional and/or
3j translational control sequences) is capable of expressing a




~.:' : '' . ' '~'
"" ~ . , . "~

2~70a8

- 38 -
DNA (or cDNA) molecule which has been cloned into the vector
and of thereby producing a polypeptide or protein.
Expression of the cloned sequences occurs when the
expression vector is introduced into an appropriate host
5 cell. If a prokaryotic expression vector is employed, then
the appropriate host cell would be any prokaryotic cell
capable of expressing the cloned sequences. Similarly, if a
eukaryotic expression vector is employed, then the
appropriate host cell would be any eukaryotic cell capable
10 of expressing the cloned sequences. Importantly, since
eukaryotic DNA may contain intervening sequences, and since
such sequences cannot be correct
ly processed in prokaryotic cells, it is preferable to
employ cDNA fr~m a cell which is capable of expressing R-
15 PTPase in order to produce a prokaryotic genomic expressionvector library. Procedures for preparing cDNA and for
producing a genomic library are disclosed by Sambrook et al.
supra) .
A DNA sequence encoding the R-PTPase of the
20 present invention, or its functional derivatives, may be
recombined with vector DNA in accordance with conventional
techniques, including blunt-ended or staggered-ended termini
for ligation, restriction enzyme digestion to provide
appropriate termini, filling in of cohesive ends as
25 appropriate, alkaline phosphatase treatment to avoid
undesirable joining, and ligation with appropriate ligases.
Technigues for such manipulations are disclosed by Sambrook
et al., supra, and are well known in the art.
A nucleic acid molecule, such as DNA, is said to
30 be "capable of expressing" a polypeptide if it contains
nucleotide sequences which contain transcriptional and
translational regulatory information and such sequences are
"operably linked" to nucleotide sequences which encode the
polypeptide. An operable linkage is a linkage in which the
35 regulatory DNA sequences and the DNA sequence sought to be




,. .. ... . . . .... .

' ', ' ' ' ~ .
' , : , ,' ' ' : ` :.


'.','.' . . ' '' , ~ , ''. ~, ' ..

V ~ V~V l' ~, J ~ V;~ ~ I U' 1~7~
~, 7003

- 39 -
expressed are connected in such a way as to permit gene
expression. The precise nature of the regulatory regions
needed for gene expression may vary from organism to
organism, but shall in general include a promoter region
5 which, in prokaryotes, contains both the promoter (which
directs the initiation of RNA transcription) as well as the
DNA sequences which, when transcribed into RNA, will signal
the initiation of protein synthesis. Such regions will
normally include those 5'-non-coding sequences involved with
$0 initiation of transcription and translation, such as the
TATA box, capping sequence, CAAT sequence, and the like.
If desired, the non-coding region 3' to the gene
sequence coding for the protein may be obtained by the
above-described methods. This region may be retained for
15 its transcriptional termination regulatory sequences, such
as termination and polyadenylation. Thus, by retaining the
3'-region naturally contiguous to the DNA sequence coding
for the protein, the transcriptional termination signals may
be provided. Where the transcriptional termination signals
20 are not satisfactorily functional in the expression host
cell, then a 3' region functional in the host cell may be
substituted.
Two DNA sequences (such as a promoter region
sequence and a R-PTPase-encoding sequence) are said to be
25 operably linked if the nature of the linkage between the two
DNA sequences does not (1) result in the introduction of a
frame-shift mutat on, (2) interfere with the ability of the
promoter region sequenoe to direct the transcription of the
R-PTPase gene sequence, or (3) interfere with the ability of
30 the R-PTPase gene seguence to be transcribed by the promoter
region sequence. A promoter region w~uld be operably linked
to a DNA sequence if the pro~oter were capable of effecting
transcription of that DNA sequence. Thus, to express the
protein, transcriptional and translational signals
35 recognized by an appropriate host are necessary.




, ,. '~


. ~ - . . ~ , . . . . .

W(~ ~/ulu~u I'~,l/U~I/U4XY:~
2~o 7~V~

- 40 -
A promoter is a double-stranded DNA or ~NA
molecule which is capable of binding RNA polymerase and
promoting the transcription of an "operably linked" nucleic
acid sequence. As used herein, a "promoter sequence" is the
sequence of the promoter which is found on that strand of
the DNA or RNA which is transcribed by the RNA polymerase.
A "promoter sequence complement~ is a nucleic acid molecule
whose sequence is the complement of a "promoter sequence."
Hence, upon extension of a primer DNA or RNA adjacent to a
10 single-stranded "promoter sequence complement" or, of a
"promoter sequence," a double-stranded molecule is created
which will contain a functional promoter, if that extension
proceeds towards the "promoter sequence" or the "promoter
sequence complement." This functional promoter will direct
15 the transcription of a nucleic acid molecule which is
operably linked to that strand of the. double-stranded
molecule which contains the "promoter sequence" (and not
that strand of the molecule which contains the "promoter
seguence complement").
Certain RNA polymerases exhibit a high specificity
for such promoters. The RNA polymerases of the
bacteriophages T7, T3, and SP-6 are etspecially well
characterized, and exhibit high promoter specificity. The
promoter sequences which are specific for each of these RNA
25 polymerases also direct the polymerase to utilize (i.e.
transcribe) only one strand of the two strands of a duplex
DNA template. The selection of which strand is transcribed
is determined by the orientation of the promoter sequence.
This selection determines the direction of transcription
30 since RNA is only polymerized enzymatically by the addition
of a nucleotide 5' phosphate to a 3' hydroxyl terminus.
- ~wo ~equences of a nucleic acid molecule are said
to be "operably linked" when they are linked to each other
in a manner which either permits both 6equences to be
35 transcribed onto the same RNA transcript, or permits an RNA

.~




. :... . . ~........................ ..
.. :

2 ~.~(, 7 ~ i)3

transcript, begun in one sequence to be extended into the
second sequence. T~us, two sequences, such as a promoter
sequence and any other ~second~ sequl~nce of DNA or RNA are
operably linked if transcription commencing in the promo~er
sequence will produce an RNA transcript of the operably
linked second 6eguence. In order to be ~operably llnked" it
is not necessary that two seguences be immediately adjacent
to one another.
Thus, as indicated above, :in order to function as
10 a promoter, a promoter sequence must be present as a double-
stranded molecule. For the purposes of the present
invention, the two strands of a functional promoter sequence
are referred to as a "transcript" strand and a
"complementary" strand. The "transcript" strand is that
15 strand of the duplex which will be transcribed by the RNA
polymerase (i.e. which serves as the template for
transcription). The "complementary" strand is the strand
which has a sequence complementary to the "transcript"
strand, and which must be present, and hybridized to the
20 "transcript" strand, in order for tr~mscription to occur.
Thus, when the "transcript" strand o e a promoter sequence is
operably linked to a second sequence, hybridization of the
"transcript" strand with the "comple~ent" strand, will, in
the presence of a polymerase, result in the transcription of
25 the "transcript" strand, and will produce an RNA transcript
using the sequence of the "transcript:" strand as a template.

The promoter sequences of the present invention
may be either prokaryotic, eukaryotic: or viral. Suitable ~`
promoters are repressible, or, ~ore preferably,
30 constitutive. Examples of suita~le ~prokaryotic promoters
include promoters capable of recognizing the T4 (Malik, S.
et al., J. Biol. Chem. ~~:1174-1181 (1984); Rosenberg, A.H.
et al., Gene 59:191-200 (1987~; ~hinedling, S. et al., J.
Molec. ~iol. 195:471-480 (1987); Hu, ~. et al., Gene 42:21-




: . , j ;. " . . : .

~7'J~
- 42 - -
30 (1986) ), T3, Sp6, and T7 (Chamberlin, M. et al., Nature
228:227-231 (1970); ~ailey, J.N. et_al , Proc. ~atl. Acad.
Sci. (U.S.A.~ 80:2814-2818 (1983); I~avanloo, P- ~
Proc. Natl. Acad. Sci. (U.S.A.~ 81:;'035-2039 (1984) )
5 polymerases; the PR and PL promoters; of bacteriophage lambda
(The Bacteriophaae Lambda, Hershey, A.D., Ed., Cold Spring
Harbor Press, Cold Spring Harbor, NY (1973); Lambda II,
Hendrix, R.W., Ed., Cold Spring Harbor Press, Cold Spring
Harbor, NY (1980)); the trD~ recA, heat shock, and lacZ
10 promoters of E. coli; the ~-amylase I~Ulmanen, I., et al., J.
Bacteriol. 162:176-182 (1985)) and t:he ~-28-specific
promoters of B. subtilis (Gilman, M.Z., et al., Gene 32:11-
20 (1984)); the promoters of the bac:~eriophages of Bacillus
(Gryczan, T.J., In: The Mel~eY~ ology of the Bacilli,
15 Academic Press, Inc., NY (lg82)); ~:reptomyces promoters
(Ward, J.M., et al., Mol. Gen. Genet~ 203:468-478 (1986));
the int promoter of bacteriophage la,~bda; the bla promoter
of the B-lactamase gene of pBR322, and the CAT promoter of
the chloramphenicol acetyl transfera~e gene of pPR325, etc.
20 Prokaryotic promoters are reviewed by Glick, B.R. (J. Ind.
crobiol. 1:277-282 (1987)); Cenatiempo, Y. (Biochimie
68:505-516 (1986)); Watson, J.D. et 3~1- (In: Molec~19F
Bioloay of the Gene, Fourth Edition, Benjamin Cummins, Menlo
Park, CA (1987)); and Gottesman, S. ~Ann. Rev. Genet.
25 18:415-442 (1984)). Preferred eukaryotic promoters include
the promoter of the mouse metallothionein I gene (Hamer, D.,
et al., J. Mol. Appl. Gen. ~:273-288 (1982)); the TK
promoter of Herpes virus (McKnight, S., Cell 31:355-365
(1982)); the SV40 early promoter (Benoist, C., et al.,
30 Nature (~ondon~ 290:304-310 (1981)); and the yeast qal4 gene
promoter (Johnston, S.A., et al., Proc. Natl. A~cad. Scis
(USAl 79:6971-6975 (1982); Silver, P~A., et al., Proc. Natl.
Acad. Sci. (USA~ 81:5951-5955 (1984)). All of the above
listed references are incorporated ~y reference herein.




.. . . . , ., .:: .: :. :
:: . . : . . . .:., . ~ - ,
; . : -
: . . : , : ~ . . - - .

... . .
.~ . - ' :: ,,
- , . . - .

20~ 70~8

- 43 -
Strong promoters are preferred. Examples of such
preferred promoters are those which recognize the T3, SP6
and ~7 polymerases, the PL promoter of bacteriophage lambda,
the recA promoter and the promoter ol the mouse
metallothionein I gene. A most preferred promoter for
eukaryotic expression of R-PT~ase is an sv40 promoter such
as that driving transcription in the pLSV vector (Livneh,
E., et al., (1986) J. Biol. Chem. 26~, 12490- 12497). The
sequences of such polymerase recognition sites are disclosed
10 by Watson, J.D. et al. (In: Molecular Bioloqy of the Gene,
Fourth Edition, Benjamin/Cu~mings Publishing Co., Inc.,
Menlo Park, CA, (1987)).

Having now generally described the invention, the
same will be more readily understood through reference to
15 the following example which is provided by way of
illustration, and is not intended to be limiting of the
present invention, unless specified.

EXA~PLE I
Isolation and Analysis of Murine R-l'TPase-~ cDNA Clones

20 1. Library screen m_
A mouse BALB/C brain cDNA li.brary in lambda gtll
(obtained from Dr. Y. Citri) was screened at relaxed
stringency (6XSSC, 5XDenhardts, 0.1% 8DS, 50 mM Tris pH 7.5,
1 mM EDTA, 0.1 mg/ml salmon sperm DNA, hybridization
25 temperature 50C) using as a probe a 2400 bp BglII-AccI
fragment representing the intracellul~r and trans-membrane
domains of the human T200 glycoprotein (Ralph, S.J. et al.,
(1987) EMB0 J. 6, 1251-1257), which had been 32P-labeled
using the random-priming method. Washing was performed at
30 50C in 6XSSC, 0.1%SDS. Out of 106 clones, 51 positives were
picked, selected and characterized by restriction enzyme
mapping. EcoRI fragments of 0.95, l.~i and 0.3 Kb isolated




~. : . : .: .: ` `

~ ~UIU~ /U:~YI~W~Yf
20~70~8


from the phage clone containing the longest insert (lambda-
lO9) were subcloned into the Bluescript KS plus and minus
vectors. A series of nested deletions were generated by
taking use of restriction sites common to the clo~ed cDNA
S fragments and the polylinker region of the plasmid vector.
The individual restriction sites used are indicated in
~igure lb. Single stranded DNA was prepared from these con-
structs, and used as a template for sequence analysis using
the dideoxynucleotide chain termination method (Seguenase,
10 United States Biochemical). All regions were sequenced on
both strands. The relative order and orientation of the
EcoRI fragments in the recombinant ph~ge was determined by
restriction mapping. To ascertain that the different EcoRI
fragments did not correspond to unrelated cDNA fragments
15 ligated together during the process of library construction,
restriction mapping was also performed on a different and
independent isolate, lambda-113.
2. Results
Brain tissue already has proven to be a rich
20 source of many types of tyrosine kinases, and recent
biochemical evidence has also indicated the existence of
multiple forms of PTPase activity (Jones, S.W- et ~1~,
(1989) J. Biol. Chem. 264, 7747-7753). In order to search
for new receptor-type PTPase, the present inventors screened
25 at low stringency a mouse brain cDNA library, using as a
hybridization probe the intracellular domain of human CD45
containing two tandem PTPase domains (Tonks, N.K. et a}.,
supra; Charbonneau, H. et al., su~ra; Ralph, S.J. et 4k,
supra). Positive clones were classified by cross-
30 hybridization and restriction mapping into several
categories, and the longest phage insert tlambda-109) -
corresponding to the most abundantly represented class was
chosen for subcloning and further analysis.
The result of the nucleotide sequence analysis is
35 shown in Figure 1. Conceptual translation of the cDNA




. . .~ . . .
~ . : . : , :


.

2~7~a8


sequence reveals the existence of a major open reading frame
of 794 amino acids, assuming that translation icnitiates at
nucleotide 259 (an in-frame stop codon is present 60
nucleotides upstream). The putative initiation methionine
5 codon is embedded in a relatively standard environment for
initiation of translation (Kozak, M., (1987) Nucl. Ac. ~es.
15, 8125-8148), and is followed by a characteristic
hydrophobic stretch of amino acids which probably function
as a signal peptide. According to the "-3,-1" rule (von
10 Heijne, G. (1986) Nucl. Ac. Res. 14, 4683-4690), residues 20
and 25 are both likely candidates to constitute the N-
terminus of the mature protein. A second hydrophobic
stretch is found between amino acids 143 and 166, and is
followed by a series of highly charged residues, consistent
15 with the stop-transfer signals found to be associated with
many membrane-spanning domains. The predicted intracellular
domain of the protein consists of two tandem repeats having
44% sequence identity between each other (residues 259-486
and 552-776). Each of these repeats display significant
20 sequence identity with the intracellular catalytic domains
of the previously described transmembrane PTPase CD45
(Ralph, S.J. et al., supra) and LAR (Streuli, M. et al.,
(1988), supra) (45% and 53% amino acid sequence identity,
respectively). In contrast, the ~MBL and GENBANK
25 databases contain no significant homology to known sequences
of the putative extracellular domain of the encoded protein.
Features of the extracellular domain include a uniquely high
content of serine and threonine residues (>32%), the absence
of cysteine residues, and the presence of 8 potential N- -
30 linked glycosylation sites.
It was concluded that the isolated cDNA encoded a
new member of the trans~embrane PTPase family having a novel
type of extracellular domain. In view of its receptor-like
structure and the likelihood that additional members of this
3~ family can be found based on the present experimental




...... .- - - ~
. ~
: ;:.: . .. .. ... . . .. , : .
, :, .. . . . .

- ,
.. . ..
- ~ : ~ : . ..
. .
.

20~70~8

-- 46 --
evidence, the name muR-PTPase-~ (m~rine Eeceptor ~rotein
tyrosine ~hosphatase-~) was chosen to desiqnate this
protein.

~P~ ~
Chromosomal Localization of thç Mouse R-P~e-~ Gene
STS/A, 020/A, CXS and OXA recombinant inbred ~RI )
mlce, and CXB ~I strains N, O, P, Q, and R were a gi~t ~rom
Dr. Jo Nilgers (The Netherlands cancer Institute). Al 1
other inbred mice were purchased fro~ the Jackson Laboratory
10 (Bar Harbor, Maine). Backcross (BC) animals were bred at
New York University with inbred progenitors obtained from `~
the Jackson Laboratory. The female parent is named first in
all crosses and Fl designations. Spleen genomic DNA from
the AXXD, AKXL, BXD, BXH and G, ~, I SWXL RI strains, and
15 from CXB, RI strains D, E, G, H, I, J, and K was purchased
from the DNA Resource at the Jackson Laboratory. For all
other mice, genomic DNA was prepared from crude liver nuclei
by a atandard sequence of protease digestion, phenol and
chloroform extraction, and et~anol precipitation. Mouse
20 genomic DNAs were subjected to Southern blotting analysis by
slight modifications of standard procedures, exactly as
described previously (Silver, J. (1985) J. Nered. 76, 436-
440). A 1.8 kb EcoRI fragment corresponding to the
intracellular phosphatase domains of R-PTPase-~, and a 0.7
25 kb SacII-EcoRI fragment corresponding to its extracellular
and transmembrane domains, were clQned into the Bluescript
KS vector, yielding plasmids plO9 and p923, respectively.
DNA restriction fragment length variants
a~sociated with the Il-la locus (interleukin-l alpha) were
30 detected by Southern blotting as described previously
(D'Eustachio, P. et al,, (1987) Im~uno~enetics 26, 339-343).
The significance of deviations from 1:1 segregation for
pairs of markers was calculated by the Bayesian method of
Silver and Buckler (Silver, J. et al,, (1986) Proc. Na~
- .




" :
" ' .


:,. ,~ - ~: . -: - - . . . -
,- - , ~ .:. - . .
. . - . . : . ., . , .. -: ... ~ . . ...... .. .... ............ . . .

. ~ : . ~ : . ... ..

-V ::~ttl~U:~U 1 ~ V~7~
2t)~7~8


Acad. Sci. USA 83, 1423-1427); Blank, R.D. et al., (1988)
Genetics 120, 1073-1083). Map distances were estimated from
recombination fractions measure in RI strain sets according
to B.A. Taylor (in: Morse, H.C. III, ed., origi~s of Inbred
5 ~ , Academic Press, New York, 1978, pp. 423-438), and
their associated 95% binomial confidence limits were
calculated according to Silver (1985, supra). Probabili~ies
o~ alternative orders of trios of markers were calculated
according to D. Bishop ((1985) Genet. Epidemiol. ~, 349-361,
10 e~uation 1). Computations were carried out on a VAX6000-410
computer.
Southern blotting analyses of genomic DNA from
inbred strains of mice revealed two useful restriction
length variants, one visualized with a probe corresponding
15 to the intracellular domain of muR-PTPase-~ ~plO9) and one
visualized with an extracellular and transmemkrane domains
probe (p923). Together, these variants allowed definition
of three allelic forms of muR-PTPase-~ among the 10 inbred
strains of mice surveyed (Table 1).
~0 TABLE 1
Restriction Fragcent Lengt~l Variants
Detected by ~uR-PTPase-~ Probes
Allele Probe Mouse Strains
plO9 p923

a 9.4 5.9+4.2 BALB/cJ
b 6.5 4.2+1.8 C57BL/6J, C57L/J, DBA/2J
c 6.5 5.9+4.2 C3H/~eJ, 020/A, AKR/J,
3D SWR/J, SJL/J, STS/A
. _
Liver genomic DNA digested with ~gI restriction
endonuclease was analyzed by Southern blotting. Fragment
sizes in kilobases ~re ~hown.




. .: ' ' '' '
.
- : .. ....... .

., . .
.

2~7008

- 48 -
In~eritance of these alleles in RI mice was scored.
Comparison of the strain distribution patterns observed for
muR-PTPase-~ (~able 2) with those previously observed for
other markers of known chromosomal location in these mice
5 indicated close linkage between the ~uR-PTPase-~ and Il-la
(Interleukin-l) loci on chromosome 2 (3 RI strains among 89 -~
examined). This degree of concordance has a probability of
less than 0.00001 of occurring as a chance event were the
loci unlinked. The observed fraction of recombinant strains
10 indicates a map distance of 0.9 cM between the loci (95%
confidence limits 0.2 - 0.6 cM).
. .




'



: .: :. .. :. .. ,.,.".. ... . . .. ; . .. ... , . - .. ....

WO 92/01050 PCI`/US91/04892
2 0 ~ 8

- 49 -

TABLE 2
Inheritance of ~uR-PTPase-~ iand Il-la DNA
~equence variants in RI stlrains of mice
AXXD strain:
1 1 1 1 1 1 1 ~ 2 2 2 2 2 2 2 2 2
1 2 3 6 ~ 8 9 o 1 2 3 4 5 6 ~3 o 1 2 3 4 5 6 7 8
Il-la D D D A D A A D D A D A D A D A D A D A D A A A
R-PTP-~ D D D A D A A D A A D A D A D A D A D A D A A A
AKXL strain: SWXL strain:
1 1 1 1 1 1 2 2 2 2 2 3 3 1 1 1 1 1
5 6 7 8 9 2 3 4 6 7 9 1 4 5 ~ 9 7 8 4 7 2 4 5 6 7
Il-la L L L A L A L L L A A L A L A, L A L S L S L L S L :
R-PTP-~ L L L A L A L L L A A L A L A L A L S L S L L S L
CXB strain: CXS strain
1 1 1 1 1
D E G H I J K N O P Q R 1 2 3 4 5 6 7 8 9 0 1 2 3 4
Il-~a C B B C B B B C C C C B T T T C C T T T C T C C T C `~
R-PTP~ C B B C B B B C B C C B T T T C C T C T C T C C T C
BXH STRAIN: BXJ strain:
1 1 1 1 1
2 3 4 6 7 8 9 0 1 2 4 9 1 2
Il-la B B H H B H B H B H H B B B
R-PTP-~ B B H H B H B H B H ~ B B B :;
RI fitrains were typed for alleles of ~uR-PTPase-~ and Il-la
by Southern blotting of TaqI-digested DNA (see Table 1 and
D'Eustachio, P. et al., Immunoaenetics 26, 339-343 (1987)).
Il-la alleles for AKXD, CXB strains D-K, and BXH mice were
disclosed in D'Eustachio et al., supra~. All RI strains are
homozygous for one of the progenitor strain alleles at each
locus; the allele is indicated by an uppercase letter
corresponding to the parent strain as ~Eollows:
A, AKR/J; B, C57BL/6J; C, BA~31c; D, DBA/2J; H, C3H/HeJ; J,
SJL/J; L, C57L/J; S, SWR/J; T, STS/A.




- . . . - . - . . , - , . .

2~ ~ 0~8

- 50 -
Following the inheritance of muR-PTPase-~, Il-la
and a (nona~outi) among progeny of reciprocal backcross
between the C57BL/6J and SWR/J strains confirmed the linkage
of muR-PTPase-~ and Il-la, and suggested an order for the
5 two genes (Table 3). Of 150 progeny, 14 were recombinant
between muR-PTPase-~ and a, and one was recombinant between
muR-PTPase-~ and Il-la. If the locus order were:
centromere-Il-l~-muR-PTPase-~-a, these results would re-
quire the occurrence of no double crossovers; alternative
10 orders require one or 14 such eventst and, evaluated
according to the method of Bishop (supra), are at least 9.5-
fold less likely. The distance between l-la and muR-
PTPase-~, 0.6 cM ~95% confidence limits: 0.1-2.4 cM),
agrees within sampling fluctuation with the distance
15 estimated from the RI strain data. Comparison of these
results with results recently obtained for Bmp-2a (Bone
morphogenic protein 2a, Dickinson, M.E. et al., (1990)
Genomics 6, 505-520) suggests that the two genes may be
closely linked, although there is no obvious structural
20 homology between them.




. . :. . , - , ,

2~7~8
- 51 -
TABL~ 3
Linkage Among ~ar~Qrs of Chromosome 2 in Backcros~ BC Progeny
A. ALLELE COMBINATIONS FROM Fl PARENT AND THE ACTUAL
5 NUMBERS OF C57BL/6J-DERIVED (b) AND SWR/J-DERIVE~ (s)
ALLELES FOUND
LOCUSP0SSI~LE ALLELE COMBIN~TION ~b ~s
.
Il-la. b s b s b s b s 76 74
1 0
R-PTP-~b s b s s b s b 77 73
ab s s b s b b s 69 81

B. NUMBERS OF PROGENY FROM EACH BACKCROSS THAT INHERITED .
EACH POSSIBLE ALLELE COMBINATION.
BACKCROSS NUMBER OF PROGENY

Fl x B 44 439 1 0 1 0 0
B x Fl 21 272 2 0 0 0 0

. j
135 14 1 0

150 progeny from BC between (C57BL/6J x SWR/J)F~ (F1) and
C57BL/6J (B) mice were typed visually for inherltance of the
25 nonagouti (a) marker and, by Southern blotting, for alleles
of the muR-PTPase-~ and Il-la loci.

~LE III ' ' '
Expression of the Murine R-P~Pase-~ RNA
30 1. Northern Analysis.
Poly A+ RNA was prepared from adult mouse tissues
and cell lines by oligo(dT) selection as described
(Vennstr~m, B. et al., (1982) ÇÇ11 28, 135-143),
fractionated (5~g per lane) on a formaldehyde-containing gel
35 and transferred to nitro~ellulose (Hybond C, Anersham) using

W() 92/01050 I'~_l/U:~YI/v~SY~
20070~8

- 52 -
standard procedures. A 32P-labelled probe was prepared by
primer extension on a single-stranded template consi6ting of
the entire lambda-109 cDNA cloned into the EcoRI site of the
Bluescript vector in the antisense orientation, using the
S Klenow fragment of DNA polymerase for elongation from an
annealed T7 primer, ~n the presence of 32p_ dATP.
Hybridization was performed at 42C in 50% formamide, 5xSsC,
25 mM KP04, 5X Denhardt's, 10 ~g/ml salmon sperm DNA, and
10% sulfate. Washing was done at 48C in O.lX SSC, 0.1%
10 SDS. Higher stringency washes (58C) of the filter did not
noticeably affect the hybridization pattern.
2. Expression of the MuEine R-PTPase-~ protein.
The entire cDNA insert from phage lambda-109 was
released as one fragment from the phage using partial EcoRI
1~ digestion, and cloned into the Bluescript KS vector. A cDNA
fragment lacking most of the untranslated leader sequence
(starting from the Sac II site at position 226; see Figure
lb) was subcloned into the SV40 promoter driven pLSV-vector
(Livneh, E., et al., (1986) J. Biol. Chem. 261, 12490-
20 12497), and the resulting plasmid DNA (pLSV-PTP-~) was
trans$ected into COS cells using the DEAE-dextran method
(Lopata, ~.A. et al., (1984) Nucl. Ac. Res. 12, 5707-5717).
The expression vector pLSV C encoding the N-terminally
truncated muR-~TPase-~ protein was used as a control in the
25 immunoprecipitation experiment.
3. Results
Poly A+ RNA from various mouse tissues was
prepared to study the expression of the muR-PTPase-~ gene.
Northern analysis (Figure 2) revealed a wide pattern of
30 expression. A 3.0 kB mRNA was present in all tissues
examined, except spleen, with brain and kidney showing the
hiahest levels of expression. An mRNA of similar size could
also be observed in the NIH-3T3 mouse fibroblast line, 2.2,
and the prepro-B lymphoid cell line, 8AF (Figure 2).
35 Shorter exposure of the Northern blot clearly showed that in




.. :. . . , -. . : :. . -

. :.. ,. . . . , ~: :.. : , . . .
. .,. .: , . - . ~ , . :
: :. :. . , . , - ,

U ~ ~JJ~J r ~ V 07~.
20~7~08

- S3 -
addition a second mRNA species of very similar size (3.2 kb)
is present in several tissues (e.g. brain) in lower amounts.
The data also suggest that, although a poly A tail and a
polyadenylation signal at the 3' end of the cDNA sequence
5 were not observed, the isolated cDNA clone (2872
nucleotides) closely matches the full length of the mRNA.
,
EXAMPLI~ IV
Transient Expression of the Murine R-PTPase-~ Protein
. , ,
1. Antibody Preparation and Immunoprecipitation.
lD Rabbits were injected with a synthetic peptide
corresponding to the predicted C-terminus of the muR-PTPase-
protein (residues 777-794) coupled to 8SA using EDCI (1-
ethyl-3- (dimethylaminopropyl)carbodiimide) as a coupling
reagent. Antigen was injected intradermally and
15 subcutaneously in an emulsion of 1 mg peptide and complete
~; Freund's adjuvant. Three booster injections were given at
; 2-3 week intervals with 0.5 mg peptide and incomplete
adjuvant. An antiserum obtained using this method was
designated "2A." Metabolic t35S]-methionine labelling, cell
20 extract preparation (60 hours after transfection) and
indirect im~unoprecipitation using protein-A-Sepharose were
performed using standard procedures (~arden, Y. et al.,
(1987) EMBO_J. 6, 3341- 3351).
2. Results
In order to determine the size of the mature
protein, we cloned the muR-PTPase-~ cDNA with the exception
of most of the untranslated leader into the pLSV vector
(Livneh, E., et al., (1986) J. Biol. Chem. 261, 12490-12497)
under the control of the SV40 promoter, yielding the
30 expression vector pLSV-PTP-~. The vector was transfected
into COS cells, and 60 hours later [35S]-methionine labelled
total cell extracts were prepared for immunoprecipitation,
using antiserum 2A.

~ .

i,

/U I US~ 'l'/ U~ Z
2~ ~0~8


As seen in Figure 3, the antiserum recognized
several bands, one of which, a diffuse band of 130 kDa
(arrow), was only present in immunoprecipitates from
transfected cells (lane 5), but not from mock-transfected
5 cells (lane 3) (transfected with pLSV without the muR-
PTPase-~ cDNA). Precipitation could be competed out by the
peptide used for immunization (lane 6).
The difference between the predicted (88 kDa) and
observed (130 kDa) molecular weights for the muR-PTPase-
~
10 protein is ascribed to its extensive glycosylation.
As an additional control for the specificity ofthe antiserum, we also transfected COS cells with a N-
truncated version of the muR-PTPase-~ cDNA (starting at
amino acid 214, and thus lacXing the. transmembrane and
15 extracellular domains) in the same vector. A new and
abundant protein with ~n apparent molecular weight of 55 kDa .
appeared in immunoprecipitates from cells transfected with
this vector, which was again competed out by the antigenic
peptide (lanes 7 and 8). The higher abundance of the
20 truncated protein as compared to the mature muR-PTPase-
~protein was a consistent observation over several
independent transfection experiments.
General Discussion for_~xamples I-IV
The Examples presented above describe the
25 identification of a novel receptor-like PTPase, R-PTPase-~,
having a broad pattern of expressionO R-PTPases are
therefore expected to have widespread functions beyond the
regulation of lymphoid cell activity, as was previously
thought based on study of CD45.
Studies using monoclonal antibodies directed
against the extracellular domain of CD45 proteins showed
that cross-linking of R-PTPases can have profound effects on
various cellular activities, although a direct effect on
PTPase enzy~atic activity re~ains to be shown. However,
35 since ligand-induced receptor cluster.irlg is a central event




.


, . .

'/U I U~ l / U~
20070a8

- 55 -
in transmembrane siqnalling by receptor tyrosine kinases
(Ullrich, A. et al., sup~), it is proposed by the inventors
that putative extracellular ligands for ~-PTPases have the
capacity to regulate the activity of R-PTPases in vivo.
In a manner analogous to that proposed for
receptor tyrosine k~nases ~PTKs), R-PTPases are proposed to
have arisen through several gene fusion events between an
ancestral PTPase domain, and domains capable of binding
extracellular ligands (Ullrich, ~. et al., Hanks, S.K. et
10 al., supra).
The variety of extracellular domains potentially
joined to PTPase domains to form receptor-like proteins are
expected to reflect the range of possible ligands able to
act by similar mechanisms. The availability of cloned R-
15 PTPases, such as those disclosed herein, will be valuable indetermining their substrate specificity and in understanding
their function and manipulating their activity.
R-PTPases might have a broad specificity directed
towards major tyrosine kinase substrates, with their
20 different extracellular domains mainly allowing for
different regulatory mechanisms responsive to different
signals in the extracellular environment. Based on this
view, they are expected to modulate the responsiveness of a
cell to those polypeptide growth factors which act through
25 receptor protein tyrosine kinases. As with PTK's, ligand
binding would lead to an activation of enzymatic activity.
Viewed in this light, R-PTPase-~ and molecules like it,
would be negative growth regulators and can be considered
potential recessive oncogenes.
For instance, deletion of portions of murine
chromosome 2, to which R-PTPase-~ map~, appears to be an
early event in the development of radiation-induced myeloid
leukemia in SJL/J mice ~Tracktenbrot, L. et al., ~1988)
Leukemia 2, 545-550), consistent with the recessive oncogene
35 notion. Furthermore, rearrangements involving human




, , ': . .. .,: , .,: , . , , ; . . ' '

v~ v~ v-~v~
20~)71~08

~ 56 -
chromosome 20 (to which the human E~~TPase-~ gene map~) have
been linked to human lymphoid leukemli.a (Mitelman, F. (ed.)
Catalog of ChroAmosome Aberrations in Human Cancer, A. Liss,
New York).
Alternatively, R-PTPase~ ay act in a manner
analogous to that proposed for the interaction between CD45
and c-lck (Oostergaard, H.L. et al , (1989) Proc. Natl.
Acad. Sci. USA 86, 8959-8963; Mustelin, T. et al., (1989
Proc. Natl. Acad. Sci. USA 86, 6302-~306~. According to
10 this view, R-P~Pase-~ would dephosphorylate negative
regulatory sites in membrane associaked PTXs which are not
receptors, and which are more widely expressed than lck
(such as, for example, the tyr527 eite in pp60C-Src).
Acting in this manner, R-PTPase-~ would be implicated in
1~ positive growth control and differentiation.
Although the inventors do not intend to be bound
by any particular theory, the high imterspecies conservation
of the catalytic domains of the varic~us receptor-PTPases
indicate an important role for these receptors in cell
20 growth control.

,EX2~L~ V,
Isolation and Characterization of ,Human R-PTPase cDNA
(Xaplan, R. et al., Proc. ~atl. Aoad.
Sci. USA 87:7000-7004 (1990))
2~ A. Materials
Restriction endonucleases and modifying enzymes
were purchases from 3Oehringer-Mannhelm or Mew England
Biolabs. Taq DNA polymerase was from Perkin-Elmer/Cetus.
The lambda gtll forward and reverse p~imers (24-mers) used
30 in the polymerase chain reactions as well as all sequencing
primers, were synthesized on an auto~lzlted DNA ~ynthesizer
(Applied Biosystems, model 380A) usinS~ either methoxy or B-
cyanoethyl phosphoramidites (House, C , et al., J. ~iol.
Chem., 262:772-777 (1987)). The lambda gtll human brainstem




. , , . - : . . ..

,
. . -
- . . .. . .
,

2~107l~8


CDNA library was obtained form the ~merican Type Culture
Collection (no. 37432). The LCA (cr)45) clone used as a
probe for screening the library was received from E.H.
Fischer (University of Washington, Seattle). All sequencing
reactions were performed using the sequenase kit (United
States Biochemical).
B. Methods
Approximately 300,000 plaques from a lambda gtll
cDNA library of 1-day-old human infant brainstem were
lO screened on duplicate nitrocellulose filters under
conditions of reduced stringency with a nick-translated LCA
probe that spanned both conserved phosphatase domains
(Charbonneau, H. et al., 1989, supra).
Hybridization was carried c~ut at 55C overnight in
15 a solution of 5x SSPE (SSPE is 10 m~ NaH2PO4, pH 7.4/0.18 M
NaCl/l mM EDTA) containing 0.25% nonfat dry milk, 0.1% SDS,
and 32P-labeled LCA probe at 106 cpm~ml. The filters were
washed three times for 20 min at 55C in 2 x SSPE/0.2% SDS
and then processed for autoradiography. This screen yielded
20 79 duplicate positives; 12 of these, showing varying degrees
of hybridization to the LCA probe, welre plaque-purified by
repetition screening with the same probe. The polymerase
chain reaction (Saiki, R.X., et al., ~;cience, 230:1350-1354
(1985)) was then used to determine thl~a sizes of the cDNA
25 inserts. The DNA templates consisted of portions of the
eluates from each pure plaque, heated at 75C for 15 min. to
release the DNA. The templates were primed with .he lambda
gtll forw?ard and reverse prim?rs. The reaction mixtures
tO.1 ml) were prepared as described (Dionne, C.A. et aL~,
30 BiQte~hni~y~ 8:190-194 (1990)). Amplification was achieved
~y performing 30 cycles, each including 1.5 min of
denaturation at 94C, 2 min of anneal:ing at 65C, and 4 min
of extension at 72C, in an automated Perkin-Elmer/Cetus DNA
thermal cycler. A portion of each sample (15 ~l) was
3j analyzed by electrophoresis through a 1% agarose gel
.




- - . . . -. ......... . , . ~ ............... .: : . .

. ~ ' . ', ?

- ~ ~ ~/ V ~ VJV
2~7~8

- 58 -
containing ethidium bromide at 1 ~g/ml (Sambrook et al.,
supra). DNA was prepared from the 4 largest clones by using
LambdaSorb (Promega) and then digested with EcoRI. The
fragments were subcloned separately into the ~QRI site of
5 M13mpl8 for sequencing. Nucleotide sequences were
determi~ed by the dideoxynucleotide chain-termination method
(Sanger, F., et al., Proc. Natl. Acad~ sci. USA, 74:5463-
5467 (1977)) using modified T7 polymerase (Tabor, s. et al.,
Proc. Natl. ~cad. Sci. US~ 84:4767- 4771 (1987)).
All computer analyses of sequence data were
performed on a Micro VAX II using programs written by
IntelliGenetics. DNA sequences were analyzed and assembled
using the GEL program. Hydrophobic analyses of proteins
were based on the algorithm of Kyte and Doolittle (Kyte, J.
15 et al., J. Mol. Biol. 157:105-132 (1982)), as implemented in
the PEP program. Protein sequence alignments were done
using the GENALIGN program (Sobel, E. et al., Nucleic Acids
Res. 14:363-374 (1985); Karlin, S. et al., Mol. Biol. Evol.
~:357-370 (1984); Needleman, S.B. et al., J. Mol. ~iol.
20 48:443-453 (1970)). Initial alignments, were done using the
Jimenez-Nontano protein alphabet (Jimenez-Montano, M. et
al., Proc. 7th Int'l. Biophysics Congress, 1981, Mexico
City).
C. Results
In an effort to identify new me~bers of the PTPase
family, 300,000 plaques from a human infant brainstem cDNA
library in lambda gtll were screened under nonstringent
conditions using a nick-translated LCA probe that spanned
both conserved phosphatase domains. Four of the initial 79
30 duplicate positives were seguenced in the entirety. ~wo
clones, 31-4 and 27-1, contained overlapping portions of the
entire coding region of a human R-PTPase (huR-PTPase) that
was designated ~ (Figure 4B). The combined lengths of
clones 31-4 and 27-1 equaled 3615 bp (Figure 4A), encoding a
35 protein of 802 amino acids (Figure 4C) and containing an




- .. . :.: - . ~ . . ..

'.,. . : ~ ' , ~' . ' . . :

~ 2/oloso PCT/US91/~92
~0~7'J~

- 59 -
additional 695 bp and 510 bp, respectively, of 5~ and 3'
untranslated region. Two of the four clones contained
portions of genes coding for two additional R-PTPases which
have been designated B and gamma (Figure 5). Like R-PTPase-
5 ~, these two proteins contain typical hydrophobictransmembrane regions and distinct extracellular domains,
indicating that they also represent separate R-PTPases.
The murine homologue of this gene was cloned and
sequenced at the same time as the human R-PTPase-~ (see
10 Examples I-IV, above). A comparison of the mouse and human
protein sequences is shown in Figure 4C. With the exception
of the extracellular domain, where some variability exists,
only 5 residues are found to differ between the two
proteins. An examination of the structure of human R-
15 PTPase-~ reveals the following features: a relatively short
extracellular domain consisting of 150 residues that
includes a hydrophobic signal peptide containing the only
cysteine in this region. There are eight potential N-
glycosylation sites, as well as a num~er of potential 0-
20 glycosylation sites (since this domain is rich in serine andthreonine). The extracellular domains of R-PTPase-~, LCA,
and LAR appear to be structurally unrelated. There is a
hydrophobic transmembrane region anchored on both sides by
charged residue6. This is followed by the two tandemly
25 repeated conserved phosphatase domains of about 235 residues
each, which are separated by 57 amino acids, typical of R-
PTPases such as LCA, LAR and the two Dro~ophila PTPases, -~
DLAR and DPTP.
Figures 5A and SB show the alignment6 of the amino
30 acids within the first and ~econd cons~rved phosphatase
domains, respectively, of LCA and R-P~Pases ~, B, and gamma.
It is readily apparent that ~mong the four R-PTPases, B and
gamma share the greatest sequence similarity. It was
reported (Hunter, T. et al. sup~) that among the sequences
35 of the conserved phosphatase domains of PTPase lB, LCA, LAR,




.... , ,. . ., .. , - . . , . .:: ...... : . . . .- ..... . . .. , . . - .


.. : :: : :: ~. . : . : : -

2~7Da~
- 60 -
DLAR and DPTP there are 29 invariant residues. While many
of these residues are also present in both phosphatase
domains of R-PTPas~-~, B, and gamma, it is interesting that
the second conserved phosphatase domains of both B and gamma
5 lack a number of these amino acids, including the two
cysteines at positions 104 and 201 in phosphatase domain 2
of LCA (see Figure 5~).
D. Discussion
The sequences of the conserved phosphatase domains
10 of the three human R-PTPases identified here (~, A, and
gamma) have been compared with one another as well as with
those of LCA, LAR, and two soluble PTPases, placental
phosphatase lB and T~cell PTPase (Table 4). The two soluble
enzymes have a sequence identity of 70%; however, when each
15 is compared with the R-PTPases (Phosphatase domains PD1 or
PD2), this nu~ber drops to 29-42%. In all cases, the
soluble PTPases showed a greater identity with PDl than with
PD2 of the R-PTPases. R-PTPase-~ appears to be most related
to ~AR, since their PDl sequences are 56% identical and
20 their PD2 sequences are 52% identical. The conserved
domains of R-PTPases B and gamma are most related to each
other, even more so than are the two soluble PTPases, B and
gamma being 75% identical in both PDl and PD2. It is
interesting that, in qeneral, the sequence relationship
25 between PDl and PD2 within any R-PTPase appears to be no
closer than that seen between different members of the
family, i.e., the identities between PDl and PD2 range from
a high of 47% for LAR to a low of 29% for R-PTPase gamma.
While the cytoplasmic domains of R-PTPase-~, B,
30 and gamma are highly conserved, the extracellular domains of
these receptors axe unrelated to one another as well as to
those of LAR and LCA. This suggests that each of thesF
receptors has its own distinct ligand. It is likely that
the binding of such ligands to the R-PTPases plays a crucial
35 role, together with growth factor receptors exhibiting




~ : .

. . V J ~ ~ U ~ V J V

20O 7~

PTRase activity, in the regulation of the level of tyrosine
phosphorylation of targets proteins involved in signal
transduction. The diversity of the R PTPases described
herein reveals the existence of a multigene family. Greater
5 understanding of structure-function relationships among
these membrane receptors will provide important insights
into the mechanisms involved in cell growth,
differentiation, and oncogenesis.




: :. :. . .- -: .. .. : : . ......... . . ..... .-
: . ., . ,....... ,., . .,. . ... : . .. ,.. . . . ~ :


. . . .. . ..

~ v ~ U 1~ 1 r~ 1 / ua~ L

21~70~8

- 62 -

Tablo 4
Identities Between Conserved Phosphatase Domains
(Perc~nt)
PTPase T cell lCA LA2 RPTP~se~ RPTPase~ RPTPase-qumma
10PTP~Ise PD1 PDl PD1 PD2 PDl PD2 PDl P02 PQl_ PD2
PTP~se la 100
T~c-ll PTPa~e 70 100
LCA PDl 37 36 100 .. .. .. .. .. .. .. .. ..
LCA P02 30 26 31 100 .. .. .. .. .. .. .. ..
LAR PDl 39 42 SO 28 100 .. .. .. .. .. .. ..
LAR PD2 29 33 42 34 45 100 .. .. .. .. .. ..
R-PTP-se-~ PDl 36 38 50 32 56 45 100 .. .. .. .. ..
R-PTP--e-q PD2 33 34 40 32 41 52 43 100 .. .. .. ..
R-PTP~se-g PDl 35 39 41 31 33 41 47 33 100 .. .. ..
R~PTPase-fi PD2 29 3D 31 30 31 34 31 37 30 100 .. ..
R~PTPase-g mna PD1 35 34 32 29 39 36 34 32 7~ 27 100 ..
iR-PTP~se-ga ~ PD2 29 29 30 28 32 36 31 34 33 75 29 100

Alignments of the conserved phosphatase dr~n ins uere cerried out as de~cribed above. The
rtgir~ns cr,npared are designated in figure 4C nd Figure 5 POspho~ph~tase dr~ in.




.~.~ .. .. . .. .. . . .. . . ... . . .

~Vo'7~)V8

- 63 -

~xpressio~ of Human R-PTPase-~ ;)rthern Blot_Analysis

: Sa~ples containing either ;20 ~g of total RNA or 2
~g of poly(A)+ ~NA were resolved in a for~aldehyde/agarose
~, 5 gel and transferred to nitrocelluloE;e. R-PTPase-~ and B-
actin probes were labeled by random priming (Sambrook ~t
al., supra). Hybridizations and waæhes were carried out at
65DC as described (Church, G., et ~1,, Proc. Natl. Acad.
Sci. USA, 81:1991- 1995 (1984)). Blots hybridized with the
lD R-PTPase-~ probe were exposed to XA~-2-x-ray film (Kodak)
with an intensifying screen for 72 h,r at -80C. Results
were obtained from the actin-probe b;lots after 15 hr under
the same conditions.
R-PTPase-~ expression was examined in various cell
15 lines and tissues (Figure 6). The results indicate the
presence of two major RNA transcripts of approximately 4.3
, and 6~3 kb, respectively. The larger: of the two species
,, appears to be ~ore prevalent in fetal tissues and in
sl parti~ularly prominent in the poly(~)+ fetal liver sample,
20 where there is also the highest relal:ive amount of the 4~3-
, kb transcript. It is possible that the different expression
,, of the twu transcripts is developmenl:ally regulated and/or a
~ result of alternative saplicing mechanisms, a feature seen
;~, with LCA (Ralph, S.J. supra). The adult brain shows
25 relatively less expression of R-PTPasi~e-~. The results
suggest that R-PTPa~e-~ is expressed to some degree
t~roughout many tissues. Murine R-PTPase-~ was also shown
. to be expressed in many tissues and cell lines and most
abundantly in brain and kidney (Sap, J., et ,al., Proc. Natl.
30 Acad. ~ci. USA, 87:6112- 6116, (1990) see also Examples III -
and IV, above).

.

..

~i


. ~. . ., :::;

W() ~2/~)10~;~) PCr/US91/0489Z
200 7l~33

-- 64 --

Chromosome Localization of the Human R-PTPase-~ Gene

Isolation, propagation, andl characterization of
parental and somatic cell hybrids using in this study have
been described (Durst, M. et al., Pro~ Natl. Acad. sci. USA
84:1070- 1074 (1987~; Ru, D-~. et al., Somatic Ce11 Mo~
Genet. 15:297-307 (1989); Juan, C-c~ et al., Proc. Natl.
Acad. Sci. US~ 85:8910-8913 (1988)). Presence of specific
human chromosomes or regions of chromosomes has been
10 confirmed by DNA hybridization using probes for genes
assigned to specific chromosome regions. Hybrid DN~s were
digested with an excess of restriction endonuclease ~a~III
or EcoRI, sized by electrophoresis in 0.8% agarose gels,
transferred to nylon filter~, and hybridized as described
15 (Durst et al., supra). The R-PTPase-l~ probe consisted of
the 3'-most 0.8 kilobases (kb) of clone 31-4 (see Figure
4B).
DNAs from 17 rodent-human somatic cell hybrids
carrying overlapping subsets of human chromosome reyions
20 representing the entire human genome were tested for
presence of the human R-PTPase-~ locu~; by Southern blot
analysis. The results (Figure 7) show that presence of the
human R-PTPase-~ locus in hybrid cells correlates only with
presence of a partial hu~an chromosome 20. The data also
25 allow a regional localization for the R-PTPase-~ locus,
since hybrids PB5-1 and AB3 are each missinq a part of the
long arm of chromosome 20 and yet retain the R-PTPase-~
locus. Thus, the human R-PTPase-~ gen~E! maps to 2Opter-
20ql2.
It has been ob~erved (Lalley, P.A. çt al.,
; Cytogenet. Cell Genet. 51:503- 532 (19~39)) that the murine
ho~ologues of all human genes which have been mapped to
human chromosome 20 map to ~ouse chromosome 2. This appears
to be true for R-PTPase-~ as well (see Example II, above).



.
, . , . ..... , ,.. ,. . . :-. -

: , . - , . : ~ . - ;: :: . -

t ~_~1 UJ~ U~ L
2 ~ 8

- 65 -
The long arm of human chromosome 20 is involved in
translocation and deletions in myeloid disorders and
neoplasms (Trent, J.M., et al., Cyto~ et. Cell Genet.,
51:533-562, (1989)). The human R-PTPase-~ locus ~ay be
5 specifically involved in deletion on 20q; in this case, it
would strengthen the possibility of it being a tumor-
suppressor gene or anti-oncoyene. Similarly in mice, in the
SJL/J strain, deletion of chromosome 2 appears to be
involved in the development of radiation-induced myeloid
lD leukemia (Trakhtenbrot, L., et al,, ~eukemia, 2:545-550,
(1988)).

The xeferences cited above are all incorporated by
reference herein, whether specifically incorporated or not.
, "
Having now fully described this invention, it will
15 be appreciated by those skilled in the art that the same can
be performed within a wide range of equivalent parameters,
concentrations, and conditions without departing from the
;~ spirit and scope of the invention and without undue
~ experimentation.
; 20 While this invention has been described in
connection with specific embodiments 1:hereof, it will be
understood that it is capable of further modifications.
!~, This application is intended to cover any variations, uses,
; or adaptations of the inventions following, in general, the
2~ principles of the invention and including such departures
from the present disclosure as come within known or
customary practice within the art to which the invention
pertains and as may be applied to the essential features
hereinbefore set forth as follows in the scope of the
30 appended claims.

,


.
. .

'

.~. :: . . .
~, . : :. ,. ,................ - -


. '' :,. . '
. . .
. , . ~ .

Representative Drawing

Sorry, the representative drawing for patent document number 2087008 was not found.

Administrative Status

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Administrative Status , Maintenance Fee  and Payment History  should be consulted.

Administrative Status

Title Date
Forecasted Issue Date Unavailable
(86) PCT Filing Date 1991-07-11
(87) PCT Publication Date 1992-01-23
(85) National Entry 1993-01-08
Examination Requested 1995-08-28
Dead Application 2007-05-03

Abandonment History

Abandonment Date Reason Reinstatement Date
2006-05-03 R30(2) - Failure to Respond
2006-07-11 FAILURE TO PAY APPLICATION MAINTENANCE FEE

Payment History

Fee Type Anniversary Year Due Date Amount Paid Paid Date
Application Fee $0.00 1993-01-08
Maintenance Fee - Application - New Act 2 1993-07-12 $100.00 1993-05-27
Registration of a document - section 124 $0.00 1993-07-13
Maintenance Fee - Application - New Act 3 1994-07-11 $100.00 1994-06-27
Maintenance Fee - Application - New Act 4 1995-07-11 $100.00 1995-06-28
Reinstatement: Failure to Pay Application Maintenance Fees $200.00 1997-01-16
Maintenance Fee - Application - New Act 5 1996-07-11 $150.00 1997-01-16
Maintenance Fee - Application - New Act 6 1997-07-11 $150.00 1997-07-04
Maintenance Fee - Application - New Act 7 1998-07-13 $150.00 1998-07-13
Maintenance Fee - Application - New Act 8 1999-07-12 $150.00 1999-06-29
Maintenance Fee - Application - New Act 9 2000-07-11 $150.00 2000-07-10
Maintenance Fee - Application - New Act 10 2001-07-11 $200.00 2001-06-28
Maintenance Fee - Application - New Act 11 2002-07-11 $200.00 2002-07-11
Maintenance Fee - Application - New Act 12 2003-07-11 $200.00 2003-06-26
Maintenance Fee - Application - New Act 13 2004-07-12 $250.00 2004-07-09
Maintenance Fee - Application - New Act 14 2005-07-11 $250.00 2005-07-06
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
NEW YORK UNIVERSITY
Past Owners on Record
SCHLESSINGER, JOSEPH
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column. To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Abstract 1995-08-17 1 46
Claims 2003-07-22 5 141
Description 2003-07-22 65 3,293
Drawings 1995-08-17 7 268
Cover Page 1995-08-17 1 29
Claims 1995-08-17 5 189
Claims 2000-11-07 4 133
Drawings 2000-11-07 13 373
Description 1995-08-17 65 3,317
Description 2000-11-07 65 3,296
Claims 2001-03-12 4 118
Claims 2004-01-07 7 182
Assignment 1993-01-08 8 266
PCT 1993-01-08 95 4,275
Prosecution-Amendment 1995-08-28 2 63
Prosecution-Amendment 1997-08-19 4 216
Prosecution-Amendment 1998-02-19 43 1,808
Prosecution-Amendment 2000-09-12 4 237
Prosecution-Amendment 2001-03-12 7 258
Prosecution-Amendment 2003-01-22 4 189
Prosecution-Amendment 2003-07-22 16 702
Fees 2000-07-10 1 44
Fees 1996-08-14 2 68
Fees 1998-07-13 1 38
Fees 1999-06-29 1 32
Fees 2004-07-09 1 33
Fees 2002-07-11 1 36
Fees 2005-07-06 1 33
Prosecution-Amendment 2005-11-03 3 166
Prosecution Correspondence 2004-01-07 10 258
Fees 1997-01-16 1 53
Fees 1995-06-28 1 41
Fees 1994-06-27 1 33
Fees 1993-05-27 1 21